1
|
Xiao T, Chen D, Peng L, Li Z, Pan W, Dong Y, Zhang J, Li M. Fluorescence-guided Surgery for Hepatocellular Carcinoma: From Clinical Practice to Laboratories. J Clin Transl Hepatol 2025; 13:216-232. [PMID: 40078203 PMCID: PMC11894393 DOI: 10.14218/jcth.2024.00375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 12/06/2024] [Accepted: 12/12/2024] [Indexed: 03/14/2025] Open
Abstract
Fluorescence navigation is a novel technique for accurately identifying hepatocellular carcinoma (HCC) lesions during hepatectomy, enabling real-time visualization. Indocyanine green-based fluorescence guidance has been commonly used to demarcate HCC lesion boundaries, but it cannot distinguish between benign and malignant liver tumors. This review focused on the clinical applications and limitations of indocyanine green, as well as recent advances in novel fluorescent probes for fluorescence-guided surgery of HCC. It covers traditional fluorescent imaging probes such as enzymes, reactive oxygen species, reactive sulfur species, and pH-sensitive probes, followed by an introduction to aggregation-induced emission probes. Aggregation-induced emission probes exhibit strong fluorescence, low background signals, excellent biocompatibility, and high photostability in the aggregate state, but show no fluorescence in dilute solutions. Design strategies for these probes may offer insights for developing novel fluorescent probes for the real-time identification and navigation of HCC during surgery.
Collapse
Affiliation(s)
- Tian Xiao
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Didi Chen
- Hubei Key Laboratory of Purification and Application of Plant Anti-Cancer Active Ingredients, Hubei University of Education, Wuhan, Hubei, China
| | - Li Peng
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhuoxia Li
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenming Pan
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuping Dong
- Beijing Key Laboratory of Construction Tailorable Advanced Functional Materials and Green Applications, School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, China
| | - Jinxiang Zhang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Min Li
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
2
|
Si J, Li C, Chen X, Zhou Q, Xue Y, Ji Y, Dong Y, Ge Z. A Near-Infrared Fluorescent Macromolecular Dye for Precise Identification of Glioblastoma Boundaries. Bioconjug Chem 2025; 36:578-587. [PMID: 40008938 DOI: 10.1021/acs.bioconjchem.5c00019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Glioblastoma (GBM) is a highly invasive tumor with poorly defined boundaries, often leaving residual tissue after surgery, which contributes to the recurrence and poor prognosis. A critical challenge in GBM treatment is the precise identification of tumor boundaries during surgery to achieve a safe and complete resection. In this study, we present a novel near-infrared fluorescent agent, IR-PEG-cRGD, that is designed to accurately delineate GBM boundaries for surgical navigation of tumor resection. IR-PEG-cRGD is successfully prepared from the cyanine dye IR-820, which is conjugated to poly(ethylene glycol) (PEG) to prolong circulation time and enhance tumor accumulation. Additionally, a glioma-targeting peptide (cRGD, cyclo(Arg-Gly-Asp-d-Phe-Cys)) is conjugated to PEG to selectively target GBM. IR-PEG-cRGD demonstrates effective targeting and enrichment in subcutaneous human-derived GBM mice models, enabling specific distinguishing of the GBM margin from the surrounding parenchyma with a high signal-to-background ratio (SBR) of 4.79. Moreover, IR-PEG-cRGD can pass across the blood-brain barrier (BBB) efficiently. These findings indicate that IR-PEG-cRGD can serve as a valuable tool for the precise intraoperative delineation of GBM boundaries, aiding in safe and complete tumor resection.
Collapse
Affiliation(s)
- Jiale Si
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Cheng Li
- Department of Geriatric General Surgery, Scientific Research Center and Precision Medical Institute, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Xin Chen
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Qinghao Zhou
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Yueming Xue
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Yuanyuan Ji
- Department of Geriatric General Surgery, Scientific Research Center and Precision Medical Institute, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yansong Dong
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Zhishen Ge
- School of Chemistry, Xi'an Key Laboratory of Sustainable Polymer Materials, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- Department of Geriatric General Surgery, Scientific Research Center and Precision Medical Institute, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| |
Collapse
|
3
|
Fang T, Dong J, Xie Z. Transformative effects of fluorescence imaging technologies on current vascular surgical practices: An updated review. SLAS Technol 2025; 32:100270. [PMID: 40086632 DOI: 10.1016/j.slast.2025.100270] [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: 10/26/2024] [Revised: 02/02/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
Fluorescence imaging technologies have revolutionized vascular surgery by enabling real-time visualization of vascular anatomy, blood circulation, and tissue perfusion, thus improving intraoperative decision-making. This review provides a comprehensive analysis of key fluorescence modalities, including Fluorescence-Guided Surgery (FGS), Near-Infrared (NIR) fluorescence imaging, and Indocyanine Green (ICG) angiography, highlighting their roles in optimizing tissue perfusion assessment, vessel patency evaluation, and identifying anatomical variations. Unlike existing literature, this review addresses critical gaps in current practices by comparing these technologies and exploring their applications across a range of vascular procedures such as peripheral vascular surgery, coronary artery bypass grafting, and oncological operations. The review further delves into the potential future directions for fluorescence imaging in vascular surgery, emphasizing emerging technologies, challenges in clinical implementation, and how these advancements can enhance surgical precision, patient outcomes, and intraoperative guidance. By synthesizing the latest developments, this review offers valuable insights into the evolving role of fluorescence imaging in vascular surgery and its potential to transform surgical practices.
Collapse
Affiliation(s)
- Tao Fang
- Department of Vascular Surgery, Yantai Mountain Hospital, Yantai 264001, China
| | - Jianxin Dong
- Department of Vascular Surgery, Yantai Mountain Hospital, Yantai 264001, China
| | - Zhilei Xie
- Department of Vascular Surgery, Yantai Mountain Hospital, Yantai 264001, China.
| |
Collapse
|
4
|
Zhang K, Li S, Li J, Zhou X, Qin Y, Wu L, Ling J. Ultra-pH-sensitive nanoplatform for precise tumor therapy. Biomaterials 2025; 314:122858. [PMID: 39366182 DOI: 10.1016/j.biomaterials.2024.122858] [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: 06/21/2024] [Revised: 09/16/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024]
Abstract
The emergence of precision cancer treatment has triggered a paradigm shift in the field of oncology, facilitating the implementation of more effective and personalized therapeutic approaches that enhance patient outcomes. The pH of the tumor microenvironment (TME) plays a pivotal role in both the initiation and progression of cancer, thus emerging as a promising focal point for precision cancer treatment. By specifically targeting the acidic conditions inherent to the tumor microenvironment, innovative therapeutic interventions have been proposed, exhibiting significant potential in augmenting treatment efficacy and ameliorating patient prognosis. The concept of ultra-pH-sensitive (UPS) nanoplatform was proposed several years ago, demonstrating exceptional pH sensitivity and an adjustable pH transition point. Subsequently, diverse UPS nanoplatforms have been actively explored for biomedical applications, enabling the loading of fluorophores, therapeutic drugs, and photosensitizers. This review aims to elucidate the design strategy and response mechanism of the UPS nanoplatform, with a specific emphasis on its applications in surgical therapy, immunotherapy, drug delivery, photodynamic therapy, and photothermal therapy. The potential and challenges of translating in the clinic on UPS nanoplatforms are finally explored. Thanks to its responsive and easily modifiable nature, the integration of multiple functional units within a UPS nanoplatform holds great promise for future advancements in tumor precision theranositcs.
Collapse
Affiliation(s)
- Ke Zhang
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, Nantong, Jiangsu, 226019, China
| | - Shijie Li
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, Nantong, Jiangsu, 226019, China
| | - Jiaying Li
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, Nantong, Jiangsu, 226019, China
| | - Xiaobo Zhou
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, Nantong, Jiangsu, 226019, China.
| | - Yuling Qin
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, Nantong, Jiangsu, 226019, China
| | - Li Wu
- Nantong Key Laboratory of Public Health and Medical Analysis, School of Public Health, Nantong University, Nantong, Jiangsu, 226019, China; School of Life Sciences, Nantong University, Nantong, Jiangsu, 226019, China.
| | - Jue Ling
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
5
|
Lee AST, Tong CMC. Novel Intraoperative Applications of Fluorescence Imaging Using Indocyanine Green in Pediatric Urology. Curr Urol Rep 2025; 26:26. [PMID: 39907934 PMCID: PMC11799058 DOI: 10.1007/s11934-025-01256-6] [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] [Accepted: 01/20/2025] [Indexed: 02/06/2025]
Abstract
PURPOSE OF REVIEW Near-infrared fluorescence imaging (NIRF) with the use of indocyanine green (ICG) has been recently adopted in pediatric urology after its well-published use in the adult population. As a powerful tool that can help delineate complex anatomy and congenital anomalies, we discuss the various applications of this imaging in minimally invasive and open surgery in pediatric urology. RECENT FINDINGS The most reported applications of ICG in pediatric urology are within minimally invasive surgery, particularly varicoceles, renal surgery such as nephrectomies and renal tumor excision, mimicking its use in adult urology. ICG has also been applied to reconstructive urology such as ureteral reconstruction, hypospadias repair and bladder exstrophy. Despite its safety and more widespread use in pediatric surgery, all published studies in pediatric urology to date have been limited to small and single-center experiences, reflecting the novel nature of this technology in this field. ICG has been shown to be safe and effective in children, particularly in those with complex anatomy and in technically challenging surgeries. Future studies should focus on standardized protocols for children and multi-center comparative studies.
Collapse
Affiliation(s)
- Albert S T Lee
- Division of Urology, Texas Children's Hospital, Houston, USA
- Department of Urology, Baylor College of Medicine, Houston, USA
| | - Ching Man Carmen Tong
- Department of Urology, University of Alabama at Birmingham, 1600 7th Avenue South Suite 318, Lowder Building, Birmingham, AL, 35233, USA.
| |
Collapse
|
6
|
Liu Y, Valji K, Monsky W, Zheng C, Yang X. Optical imaging guidance in oncologic surgery and interventional oncology. Pharmacol Res 2025; 212:107612. [PMID: 39826822 DOI: 10.1016/j.phrs.2025.107612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Over recent decades, optical imaging (OI) has become an integral part of medical imaging, offering significant advantages over other modalities, such as computed tomography (CT) and magnetic resonance imaging (MRI). OI is distinguished by its real-time imaging capability, cost-effectiveness, portability, absence of ionizing radiation, and high patient acceptability. The introduction of advanced optical dyes (including FDA-approved agents like indocyanine green, Cytalux, and Gleolan) has greatly enhanced its clinical utility. OI has shown clear benefits in the management of patients with cancer, originally by open surgery and now extending to minimally invasive, image-guided interventional procedures. This review highlights recent developments in OI for oncology, emphasizing its benefits for clinicians in guiding surgical and interventional procedures.
Collapse
Affiliation(s)
- Yiming Liu
- Image-Guided Bio-Molecular Intervention Research and Division of Interventional Radiology, Department of Radiology, University of Washington School of Medicine, Seattle, USA; Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Karim Valji
- Image-Guided Bio-Molecular Intervention Research and Division of Interventional Radiology, Department of Radiology, University of Washington School of Medicine, Seattle, USA
| | - Wayne Monsky
- Image-Guided Bio-Molecular Intervention Research and Division of Interventional Radiology, Department of Radiology, University of Washington School of Medicine, Seattle, USA
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaoming Yang
- Image-Guided Bio-Molecular Intervention Research and Division of Interventional Radiology, Department of Radiology, University of Washington School of Medicine, Seattle, USA.
| |
Collapse
|
7
|
Wu X, Deng Y, Wang R, Kim H, Kim G, Xu Y, Hong KT, Lee JS, Hu JJ, Liang G, Yoon J. Rational Design of an Activatable Near-Infrared Fluorogenic Platform for In Vivo Orthotopic Tumor Imaging and Resection. Angew Chem Int Ed Engl 2025; 64:e202416877. [PMID: 39449191 DOI: 10.1002/anie.202416877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 10/26/2024]
Abstract
Rational and effective design of a universal near-infrared (NIR) light-absorbed platform employed to prepare diverse activatable NIR fluorogenic probes for in vivo imaging and the imaging-guided tumor resection remains less exploited but highly meaningful. Herein, mandelic acid with a core structure of 4-hydroxylbenzyl alcohol to link recognition unit, a fluorophore and a quencher was employed to prepare activatable probes. We exemplified ester as carboxylesterase (CE)-recognized unit, ferrocene as quencher and phenothiazinium as NIR fluorophore to afford fluorogenic probes termed NBS-Fe-CE and NBS-C-Fe-CE. These probes enabled the conversion toward CE with significant fluorescence increases and successfully discriminate CE activity in cells. NIR light enhances the tumor penetration and enable imaging-guided orthotopic tumor resection. This specific case demonstrated that this platform can be effectively used to construct diverse NIR probes for imaging analytes in biological systems.
Collapse
Affiliation(s)
- Xiaofeng Wu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yu Deng
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Rui Wang
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03706, Republic of Korea
| | - Heejeong Kim
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03706, Republic of Korea
| | - Gyoungmi Kim
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03706, Republic of Korea
| | - Ying Xu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Kyung Tae Hong
- Bio-Med Program, KIST-School UST, Hwarang-ro 14 gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Jun-Seok Lee
- Department of Pharmacology, Korea University College of Medicine, 73 Goryeodae-ro, Seongbuk-gu, Seoul, 02841, Korea
| | - Jing-Jing Hu
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Gaolin Liang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03706, Republic of Korea
| |
Collapse
|
8
|
Spring BQ, Watanabe K, Ichikawa M, Mallidi S, Matsudaira T, Timerman D, Swain JWR, Mai Z, Wakimoto H, Hasan T. Red light-activated depletion of drug-refractory glioblastoma stem cells and chemosensitization of an acquired-resistant mesenchymal phenotype. Photochem Photobiol 2025; 101:215-229. [PMID: 38922889 PMCID: PMC11664018 DOI: 10.1111/php.13985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024]
Abstract
Glioblastoma stem cells (GSCs) are potent tumor initiators resistant to radiochemotherapy, and this subpopulation is hypothesized to re-populate the tumor milieu due to selection following conventional therapies. Here, we show that 5-aminolevulinic acid (ALA) treatment-a pro-fluorophore used for fluorescence-guided cancer surgery-leads to elevated levels of fluorophore conversion in patient-derived GSC cultures, and subsequent red light-activation induces apoptosis in both intrinsically temozolomide chemotherapy-sensitive and -resistant GSC phenotypes. Red light irradiation of ALA-treated cultures also exhibits the ability to target mesenchymal GSCs (Mes-GSCs) with induced temozolomide resistance. Furthermore, sub-lethal light doses restore Mes-GSC sensitivity to temozolomide, abrogating GSC-acquired chemoresistance. These results suggest that ALA is not only useful for fluorescence-guided glioblastoma tumor resection, but that it also facilitates a GSC drug-resistance agnostic, red light-activated modality to mop up the surgical margins and prime subsequent chemotherapy.
Collapse
Affiliation(s)
- Bryan Q. Spring
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Physics, Northeastern University, Boston, MA 02115, USA
| | - Kohei Watanabe
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Healthcare Optics Research Laboratory, Canon USA, Inc., Cambridge MA 02139, USA
| | - Megumi Ichikawa
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Srivalleesha Mallidi
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Tatsuyuki Matsudaira
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Dmitriy Timerman
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Joseph W. R. Swain
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Zhiming Mai
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Hiroaki Wakimoto
- Brain Tumor Research Center and Molecular Neurosurgery Laboratory, Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
9
|
Joshi S, Moody A, Budthapa P, Gurung A, Gautam R, Sanjel P, Gupta A, Aryal SP, Parajuli N, Bhattarai N. Advances in Natural-Product-Based Fluorescent Agents and Synthetic Analogues for Analytical and Biomedical Applications. Bioengineering (Basel) 2024; 11:1292. [PMID: 39768110 PMCID: PMC11727039 DOI: 10.3390/bioengineering11121292] [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: 11/19/2024] [Revised: 12/09/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025] Open
Abstract
Fluorescence is a remarkable property exhibited by many chemical compounds and biomolecules. Fluorescence has revolutionized analytical and biomedical sciences due to its wide-ranging applications in analytical and diagnostic tools of biological and environmental importance. Fluorescent molecules are frequently employed in drug delivery, optical sensing, cellular imaging, and biomarker discovery. Cancer is a global challenge and fluorescence agents can function as diagnostic as well as monitoring tools, both during early tumor progression and treatment monitoring. Many fluorescent compounds can be found in their natural form, but recent developments in synthetic chemistry and molecular biology have allowed us to synthesize and tune fluorescent molecules that would not otherwise exist in nature. Naturally derived fluorescent compounds are generally more biocompatible and environmentally friendly. They can also be modified in cost-effective and target-specific ways with the help of synthetic tools. Understanding their unique chemical structures and photophysical properties is key to harnessing their full potential in biomedical and analytical research. As drug discovery efforts require the rigorous characterization of pharmacokinetics and pharmacodynamics, fluorescence-based detection accelerates the understanding of drug interactions via in vitro and in vivo assays. Herein, we provide a review of natural products and synthetic analogs that exhibit fluorescence properties and can be used as probes, detailing their photophysical properties. We have also provided some insights into the relationships between chemical structures and fluorescent properties. Finally, we have discussed the applications of fluorescent compounds in biomedical science, mainly in the study of tumor and cancer cells and analytical research, highlighting their pivotal role in advancing drug delivery, biomarkers, cell imaging, biosensing technologies, and as targeting ligands in the diagnosis of tumors.
Collapse
Affiliation(s)
- Soniya Joshi
- Central Department of Chemistry, Tribhuvan University, Kathmandu 44618, Nepal; (S.J.); (P.B.); (A.G.); (R.G.); (P.S.)
| | - Alexis Moody
- Department of Chemical, Biological, and Bioengineering, North Carolina A&T State University, Greensboro, NC 27411, USA;
| | - Padamlal Budthapa
- Central Department of Chemistry, Tribhuvan University, Kathmandu 44618, Nepal; (S.J.); (P.B.); (A.G.); (R.G.); (P.S.)
| | - Anita Gurung
- Central Department of Chemistry, Tribhuvan University, Kathmandu 44618, Nepal; (S.J.); (P.B.); (A.G.); (R.G.); (P.S.)
| | - Rachana Gautam
- Central Department of Chemistry, Tribhuvan University, Kathmandu 44618, Nepal; (S.J.); (P.B.); (A.G.); (R.G.); (P.S.)
| | - Prabha Sanjel
- Central Department of Chemistry, Tribhuvan University, Kathmandu 44618, Nepal; (S.J.); (P.B.); (A.G.); (R.G.); (P.S.)
| | - Aakash Gupta
- Department of Biomedical Engineering, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA;
| | - Surya P. Aryal
- Department of Chemistry, University of Kentucky, Lexington, KY 40506, USA;
| | - Niranjan Parajuli
- Central Department of Chemistry, Tribhuvan University, Kathmandu 44618, Nepal; (S.J.); (P.B.); (A.G.); (R.G.); (P.S.)
| | - Narayan Bhattarai
- Department of Chemical, Biological, and Bioengineering, North Carolina A&T State University, Greensboro, NC 27411, USA;
| |
Collapse
|
10
|
Xu W, Jian D, Yang H, Wang W, Ding Y. Aggregation-induced emission: Application in diagnosis and therapy of hepatocellular carcinoma. Biosens Bioelectron 2024; 266:116722. [PMID: 39232431 DOI: 10.1016/j.bios.2024.116722] [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: 04/30/2024] [Revised: 08/24/2024] [Accepted: 08/28/2024] [Indexed: 09/06/2024]
Abstract
Hepatocellular carcinoma (HCC) is a serious health issue due to its low early diagnosis rate, resistance to chemotherapy, and poor five-year survival rate. Therefore, it is crucial to explore novel diagnostic and therapeutic approaches tailored to the characteristics of HCC. Aggregation-induced emission (AIE) is a phenomenon where the luminescence of certain molecules, typically non-luminescent or weakly luminescent in solution, is significantly enhanced upon aggregation. AIE has been extensively applied in bioimaging, biosensors, and therapy. Fluorophore materials based on AIE (AIEgens) have a wide range of application scenarios and potential for clinical translation. This review focuses on recent advances in AIE-based strategies for diagnosing and treating HCC. First, the specific functional mechanism of AIE is described. Next, we summarize recent progress in the application of AIE for multimodal imaging, biosensor detection, and phototherapy. Finally, prospects and challenges for the AIE-based application in the diagnosis and therapy of HCC are discussed.
Collapse
Affiliation(s)
- Wenjing Xu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; The Second Affiliated Hospital of Zhejiang University, Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; The Second Affiliated Hospital of Zhejiang University, Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease, Zhejiang University, Hangzhou, Zhejiang, 310009, China; The Second Affiliated Hospital of Zhejiang University Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Danfeng Jian
- MOE Key Laboratory of Advanced Textile Materials & Manufacturing Technology, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Huang Yang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; The Second Affiliated Hospital of Zhejiang University, Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; The Second Affiliated Hospital of Zhejiang University, Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease, Zhejiang University, Hangzhou, Zhejiang, 310009, China; The Second Affiliated Hospital of Zhejiang University Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310009, China; MOE Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| | - Weili Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; The Second Affiliated Hospital of Zhejiang University, Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; The Second Affiliated Hospital of Zhejiang University, Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease, Zhejiang University, Hangzhou, Zhejiang, 310009, China; The Second Affiliated Hospital of Zhejiang University Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; The Second Affiliated Hospital of Zhejiang University, Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; The Second Affiliated Hospital of Zhejiang University, Research Center of Diagnosis and Treatment Technology for Hepatocellular Carcinoma of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; Clinical Medicine Innovation Center of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Disease, Zhejiang University, Hangzhou, Zhejiang, 310009, China; The Second Affiliated Hospital of Zhejiang University Clinical Research Center of Hepatobiliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310009, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
11
|
Khalid A, Tomljenovic-Hanic S. Emerging Fluorescent Nanoparticles for Non-Invasive Bioimaging. Molecules 2024; 29:5594. [PMID: 39683753 DOI: 10.3390/molecules29235594] [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: 10/31/2024] [Revised: 11/19/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Fluorescence-based techniques have great potential in the field of bioimaging and could bring tremendous progress in microbiology and biomedicine. The most essential element in these techniques is fluorescent nanomaterials. The use of fluorescent nanoparticles as contrast agents for bioimaging is a large topic to cover. The purpose of this mini-review is to give the reader an overview of biocompatible and biodegradable fluorescent nanoparticles that are emerging nanomaterials for use in fluorescent bioimaging. In addition to the biocompatibility of these nanomaterials, biodegradability is considered a necessity for short-term sustainable bioimaging. Firstly, the main requirements for bioimaging are raised, and a few existing fluorescent nanoprobes are discussed. Secondly, a few inert biocompatible fluorescent nanomaterials for long-term bioimaging that have been, to some extent, demonstrated as fluorescent probes are reviewed. Finally, a few biocompatible and biodegradable nanomaterials for short-term bioimaging that are evolving for bioimaging applications are discussed. Together, these advancements signal a transformative leap toward sustainability and functionality in biomedical imaging.
Collapse
Affiliation(s)
- Asma Khalid
- School of Physics, University of Melbourne, Parkville, VIC 3010, Australia
- School of Science, RMIT University, Melbourne, VIC 3001, Australia
| | | |
Collapse
|
12
|
Chen X, Jiang Y, Liu J, Tian Y, Deng Y, Li X, Wu W, Zhang R, Deng Y. Suppressing ROS Production of AIE Nanoprobes by Simple Matrices Optimization for CNS Cell Observation and Minimized Influence of Cytoskeleton Morphology. CHEMICAL & BIOMEDICAL IMAGING 2024; 2:775-783. [PMID: 39610462 PMCID: PMC11600148 DOI: 10.1021/cbmi.4c00061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 11/30/2024]
Abstract
The visualization of the central nervous system (CNS) has proposed stringent criteria for fluorescent probes, as the inevitable production of reactive oxygen species (ROS) or heat generated from most photoluminescent probes upon excitation can disturb the normal status of relatively delicate CNS cells. In this work, a red-emitting fluorogen with aggregation-induced emission (AIE) characteristics, known as DTF, was chosen as the model fluorogen to investigate whether the side effects of ROS and heat could be suppressed through easy-to-operate processes. Specifically, DTF was encapsulated with different amphiphilic matrices to yield AIE nanoprobes, and their photoluminescent properties, ROS production, and photothermal conversion rates were examined. BSA@DTF NPs possessed 1.3-fold brightness compared to that of DSPE-PEG@DTF NPs and F127@DTF NPs but its ROS generation efficiency is markedly decreased to only 2.4% of that produced by F127@DTF NPs. Meanwhile, BSA@DTF NPs showed a negligible photothermal effect. These features make BSA@DTF NPs favorable for long-term live cell imaging, particularly for fluorescent imaging of CNS cells. BSA@DTF NPs were able to sustain the normal state of HT-22 neuronal cells with continuous illumination for at least 25 min, and they also preserved the cytoskeleton of microglia BV-2 cells as the untreated control group. This work represents a successful but easy-to-operate process to suppress the ROS generation of red-emissive AIEgen, and it highlights the importance of minimizing the ROS generation of the fluorescent probes, particularly in the application of long-term imaging of CNS cells.
Collapse
Affiliation(s)
- Xiaotong Chen
- Institute
of Engineering Medicine, School of Medical Technology, Beijing Key
Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing, 100081 P. R. China
| | - Yajing Jiang
- Department
of Chemistry, Institute of Molecular Aggregation Science, Tianjin University, Tianjin, 300072 P. R. China
| | - Jiaxin Liu
- Department
of Chemistry, Institute of Molecular Aggregation Science, Tianjin University, Tianjin, 300072 P. R. China
| | - Yu Tian
- Department
of Chemistry, Institute of Molecular Aggregation Science, Tianjin University, Tianjin, 300072 P. R. China
| | - Yifan Deng
- Institute
of Engineering Medicine, School of Medical Technology, Beijing Key
Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing, 100081 P. R. China
| | - Xiaoqiong Li
- Institute
of Engineering Medicine, School of Medical Technology, Beijing Key
Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing, 100081 P. R. China
| | - Wenbo Wu
- Department
of Chemistry, Institute of Molecular Aggregation Science, Tianjin University, Tianjin, 300072 P. R. China
| | - Ruoyu Zhang
- Institute
of Engineering Medicine, School of Medical Technology, Beijing Key
Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing, 100081 P. R. China
| | - Yulin Deng
- Institute
of Engineering Medicine, School of Medical Technology, Beijing Key
Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing, 100081 P. R. China
| |
Collapse
|
13
|
Gariglio G, Bendova K, Hermann M, Olafsdottir A, Sosabowski JK, Petrik M, von Guggenberg E, Decristoforo C. Comparison of Two Chelator Scaffolds as Basis for Cholecystokinin-2 Receptor Targeting Bimodal Imaging Probes. Pharmaceuticals (Basel) 2024; 17:1569. [PMID: 39770411 PMCID: PMC11676163 DOI: 10.3390/ph17121569] [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: 10/28/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Dual-modality probes, combining positron emission tomography (PET) with fluorescence imaging (FI) capabilities in a single molecule, are of high relevance for the accurate staging and guided resection of tumours. We herein present a pair of candidates targeting the cholecystokinin-2 receptor (CCK2R), namely [68Ga]Ga-CyTMG and [68Ga]Ga-CyFMG. In these probes, the SulfoCy5.5 fluorophore and two units of a CCK2R-binding motif are coupled to the chelator acting as a core scaffold, triazacyclononane-phosphinic acid (TRAP), and Fusarinine C (FSC), respectively. Using this approach, we investigated the influence of these chelators on the final properties. Methods: The synthetic strategy to both precursors was based on the stoichiometric conjugation of the components via click chemistry. The characterization in vitro included the evaluation of the CCK2R affinity and internalization in A431-CCK2R cells. Ex vivo biodistribution as well as PET and FI studies were performed in xenografted mice. Results: 68Ga labelling was accomplished with high radiochemical yield and purity for both precursors. A CCK2R affinity in the subnanomolar range of the conjugates and a receptor-specific uptake of the radioligands in cells were observed. In A431-CCK2R/A431-mock xenografted mice, the investigated compounds showed specific accumulation in the tumours and reduced off-target uptake compared to a previously developed compound. Higher accumulation and prolonged retention in the kidneys were observed for [68Ga]Ga-CyTMG when compared to [68Ga]Ga-CyFMG. Conclusions: Despite the promising targeting properties observed, further probe optimization is required to achieve enhanced imaging contrast at early timepoints. Additionally, the results indicate a distinct influence of the chelators in terms of renal accumulation and retention.
Collapse
Affiliation(s)
- Giacomo Gariglio
- Department of Nuclear Medicine, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Katerina Bendova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic
| | - Martin Hermann
- Department of Anesthesiology and Critical Care Medicine, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Asta Olafsdottir
- Perceptive Discovery, Hammersmith Hospital, Imperial College London, London W12 0NN, UK
| | - Jane K. Sosabowski
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London E1 4NS, UK
| | - Milos Petrik
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 77900 Olomouc, Czech Republic
- Czech Advanced Technology and Research Institute, Palacky University, 77900 Olomouc, Czech Republic
- Laboratory of Experimental Medicine, University Hospital, 77900 Olomouc, Czech Republic
| | | | - Clemens Decristoforo
- Department of Nuclear Medicine, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
14
|
Tong J, Wang Z, Zhang J, Gao R, Liu X, Liao Y, Guo X, Wei Y. Advanced Applications of Nanomaterials in Atherosclerosis Diagnosis and Treatment: Challenges and Future Prospects. ACS APPLIED MATERIALS & INTERFACES 2024; 16:58072-58099. [PMID: 39432384 DOI: 10.1021/acsami.4c13607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Atherosclerosis-induced coronary artery disease is a major cause of cardiovascular mortality. Clinically, conservative treatment strategies for atherosclerosis still focus on lifestyle interventions and the use of lipid-lowering and anticoagulant medications. Despite achieving some therapeutic effects, these approaches are limited by low bioavailability, long intervention periods, and significant side effects. With the advancement of nanotechnology, nanomaterials have demonstrated extraordinary potential in the biomedical field. Their excellent biocompatibility, surface modifiability, and high targeting capability not only enable efficient diagnosis of plaque progression but also allow precise drug delivery within atherosclerotic plaques, significantly enhancing drug bioavailability and reducing systemic side effects. Here, we systematically review the current research progress of nanomaterials in the field of atherosclerosis to summarize not only the types of nanomaterials but also their applications in both the diagnosis and treatment of atherosclerosis. Notably, in the context of plaque therapy, we provide a comprehensive overview of current nanomaterial applications based on their targeted therapeutic systems for different cell types within plaques. Additionally, we address the persistent challenge of clinical translation of nanomaterials by summarizing current issues and providing directions for innovation and improvement in nanomaterial design. Overall, we believe that this review systematically summarizes the applications and challenges of biomedical nanomaterials in atherosclerosis diagnosis and therapy, thereby offering insights and references for the development of therapeutic materials for atherosclerosis.
Collapse
Affiliation(s)
- Junran Tong
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhiwen Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiahui Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ran Gao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiangfei Liu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Yuhan Liao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaopeng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yumiao Wei
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
15
|
Kim H, Liu M, Choi Y. Quenched Zwitterionic Cyclic Arg-Gly-Asp-Containing Pentapeptide Probe for Real-Time Brain Tumor Imaging. Pharmaceutics 2024; 16:1034. [PMID: 39204379 PMCID: PMC11360280 DOI: 10.3390/pharmaceutics16081034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
The efficacy of glioblastoma treatment is closely associated with complete tumor resection. However, conventional surgical techniques often result in incomplete removal, leading to poor prognosis. A major challenge is the accurate delineation of tumor margins from healthy tissues. Imaging-guided surgery, particularly using fluorescent probes, is a promising solution for intraoperative guidance. The recently developed 'always-on' types of targeted fluorescence probes generate signals irrespective of their presence in tumor cells or in blood circulation, hampering their effectiveness. Here, we propose a novel activatable fluorescence imaging probe, Q-cRGD, that targets glioma cells via the specific binding of the cyclic Arg-Gly Asp-containing pentapeptide (cRGD) to integrins. The Q-cRGD probe was synthesized by conjugating a near-infrared (NIR) dye to a tryptophan quencher via a disulfide linkage, including a cRGD-targeting ligand. This activatable probe remained inactive until the redox-responsive cleavage of the disulfide linkage occurred within the target cell. The zwitterionic nature of NIR dyes minimizes nonspecific interactions with serum proteins, thereby enhancing the tumor-to-background signal ratio (TBR). An in vivo fluorescence imaging study demonstrated a TBR value of 2.65 within 3 h of the intravenous injection of Q-cRGD, confirming its potential utility in imaging-guided brain cancer surgery.
Collapse
Affiliation(s)
| | | | - Yongdoo Choi
- Division of Technology Convergence, National Cancer Center, 323 Ilsan-ro, Goyang 10408, Republic of Korea; (H.K.); (M.L.)
| |
Collapse
|
16
|
Chow CY, King GF. Shining a Light on Venom-Peptide Receptors: Venom Peptides as Targeted Agents for In Vivo Molecular Imaging. Toxins (Basel) 2024; 16:307. [PMID: 39057947 PMCID: PMC11281729 DOI: 10.3390/toxins16070307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Molecular imaging has revolutionised the field of biomedical research by providing a non-invasive means to visualise and understand biochemical processes within living organisms. Optical fluorescent imaging in particular allows researchers to gain valuable insights into the dynamic behaviour of a target of interest in real time. Ion channels play a fundamental role in cellular signalling, and they are implicated in diverse pathological conditions, making them an attractive target in the field of molecular imaging. Many venom peptides exhibit exquisite selectivity and potency towards ion channels, rendering them ideal agents for molecular imaging applications. In this review, we illustrate the use of fluorescently-labelled venom peptides for disease diagnostics and intraoperative imaging of brain tumours and peripheral nerves. Finally, we address challenges for the development and clinical translation of venom peptides as nerve-targeted imaging agents.
Collapse
Affiliation(s)
- Chun Yuen Chow
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
- Australia Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Glenn F. King
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
- Australia Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, St. Lucia, QLD 4072, Australia
| |
Collapse
|
17
|
Wagner P, Levine EA, Kim AC, Shen P, Fleming ND, Westin SN, Berry LK, Karakousis GC, Tanyi JL, Olson MT, Madajewski B, Ostrander B, Krishnan K, Balch CM, Bartlett DL. Detection of Residual Peritoneal Metastases Following Cytoreductive Surgery Using Pegsitacianine, a pH-Sensitive Imaging Agent: Final Results from a Phase II Study. Ann Surg Oncol 2024; 31:4726-4734. [PMID: 38622456 DOI: 10.1245/s10434-024-15165-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/25/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND For patients with peritoneal carcinomatosis, extent of disease and completeness of cytoreductive surgery (CRS) are major prognostic factors for long-term survival. Assessment of these factors could be improved using imaging agents. Pegsitacianine is a pH-sensitive polymeric micelle conjugated to the fluorophore indocyanine green. The micelle disassembles in acidic microenvironments, such as tumors, resulting in localized fluorescence unmasking. We assessed the utility of pegsitacianine in detecting residual disease following CRS. PATIENTS AND METHODS NCT04950166 was a phase II, non-randomized, open-label, multicenter US study. Patients eligible for CRS were administered an intravenous dose of pegsitacianine at 1 mg/kg 24-72 h before surgery. Following CRS, the peritoneal cavity was reexamined under near-infrared (NIR) illumination to evaluate for fluorescent tissue. Fluorescent tissue identified was excised and evaluated by histopathology. The primary outcome was the rate of clinically significant events (CSE), defined as detection of histologically confirmed residual disease excised with pegsitacianine or a revision in the assessment of completeness of CRS. Secondary outcomes included acceptable safety and pegsitacianine performance. RESULTS A total of 53 patients were screened, 50 enrolled, and 40 were evaluable for CSE across six primary tumor types. Residual disease was detected with pegsitacianine in 20 of 40 (50%) patients. Pegsitacianine showed high sensitivity and was well tolerated with no serious adverse events (SAEs). Transient treatment-related, non-anaphylactic infusion reactions occurred in 28% of patients. CONCLUSIONS Pegsitacianine was well tolerated and facilitated the recognition of occult residual disease following CRS. The high rate of residual disease detected suggests that the use of pegsitacianine augmented surgeon assessment and performance during CRS.
Collapse
Affiliation(s)
- Patrick Wagner
- Department of Surgical Oncology, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA
| | - Edward A Levine
- Department of Surgical Oncology, Atrium Health Wake Forest Baptist, Wake Forest University, Winston-Salem, NC, USA
| | - Alex C Kim
- Division of Surgical Oncology, The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH, USA
| | - Perry Shen
- Department of Surgical Oncology, Atrium Health Wake Forest Baptist, Wake Forest University, Winston-Salem, NC, USA
| | - Nicole D Fleming
- Department of Gynecologic Oncology and Reproductive Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Shannon N Westin
- Department of Gynecologic Oncology and Reproductive Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Laurel K Berry
- Department of Gynecologic Oncology, Atrium Health Wake Forest Baptist, Wake Forest University, Winston-Salem, NC, USA
| | - Giorgos C Karakousis
- Division of Endocrine and Oncologic Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Janos L Tanyi
- Department of Obstetrics and Gynecology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | | - Charles M Balch
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - David L Bartlett
- Department of Surgical Oncology, Allegheny Health Network Cancer Institute, Pittsburgh, PA, USA.
| |
Collapse
|
18
|
Sun W, Zhang L, Xing L, He Z, Zhang Y, Gao F. Projected algebraic reconstruction technique-network for high-fidelity diffuse fluorescence tomography reconstruction. JOURNAL OF THE OPTICAL SOCIETY OF AMERICA. A, OPTICS, IMAGE SCIENCE, AND VISION 2024; 41:988-999. [PMID: 38856406 DOI: 10.1364/josaa.517742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/01/2024] [Indexed: 06/11/2024]
Abstract
We propose a model-driven projected algebraic reconstruction technique (PART)-network (PART-Net) that leverages the advantages of the traditional model-based method and the neural network to improve the imaging quality of diffuse fluorescence tomography. In this algorithm, nonnegative prior information is incorporated into the ART iteration process to better guide the optimization process, and thereby improve imaging quality. On this basis, PART in conjunction with a residual convolutional neural network is further proposed to obtain high-fidelity image reconstruction. The numerical simulation results demonstrate that the PART-Net algorithm effectively improves noise robustness and reconstruction accuracy by at least 1-2 times and exhibits superiority in spatial resolution and quantification, especially for a small-sized target (r=2m m), compared with the traditional ART algorithm. Furthermore, the phantom and in vivo experiments verify the effectiveness of the PART-Net, suggesting strong generalization capability and a great potential for practical applications.
Collapse
|
19
|
Zhou X, Cai Q, Zhao S, Ling F, Xiang G, Li L, Wang Y, Li Y, Tang X. CDs-ICG@BSA nanoparticles for excellent phototherapy and in situ bioimaging. Talanta 2024; 271:125661. [PMID: 38219322 DOI: 10.1016/j.talanta.2024.125661] [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: 08/28/2023] [Revised: 01/07/2024] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
For the diagnosis and treatment of cancer, a great challenge is the fabrication of straightforward, non-toxic, multifunctional green nanomaterials. In this study, carbon quantum dots self-assembled with indocyanine green dye at bovine serum albumin for phototherapy and in situ bioimaging are produced by a flexible hydrothermal method. We find that the synthesized nanoparticles have high tumor photothermal therapeutic activity when exposed to 808 nm light, with a photothermal conversion efficiency up to 61 %. The phototoxicity study revealed the excellent phototherapy of the nanoparticles mainly arises from photothermal therapeutic effect other than photodynamic therapy effect. Simultaneously, it allows biological imaging in the visible and near-infrared ranges because of the significant absorption at 365 nm and 840 nm. The current work offers a simple, environmentally friendly, and reasonable method for developing photothermal drugs with a high photothermal conversion efficiency in the near-infrared region, as well as good biosafety for multifunctional nanomaterials for bioimaging tumor diagnosis and direct phototherapy.
Collapse
Affiliation(s)
- Xianju Zhou
- School of Science, Chongqing University of Posts and Telecommunications, Chongqing, 400065, PR China.
| | - Qingchi Cai
- School of Science, Chongqing University of Posts and Telecommunications, Chongqing, 400065, PR China
| | - Shouchun Zhao
- School of Science, Chongqing University of Posts and Telecommunications, Chongqing, 400065, PR China
| | - Faling Ling
- School of Science, Chongqing University of Posts and Telecommunications, Chongqing, 400065, PR China
| | - Guotao Xiang
- School of Science, Chongqing University of Posts and Telecommunications, Chongqing, 400065, PR China
| | - Li Li
- School of Science, Chongqing University of Posts and Telecommunications, Chongqing, 400065, PR China.
| | - Yongjie Wang
- School of Science, Chongqing University of Posts and Telecommunications, Chongqing, 400065, PR China
| | - Yanhong Li
- School of Science, Chongqing University of Posts and Telecommunications, Chongqing, 400065, PR China
| | - Xiao Tang
- School of Science, Chongqing University of Posts and Telecommunications, Chongqing, 400065, PR China
| |
Collapse
|
20
|
Gonzales J, Adilbay D, de Souza Franca PD, Artschwager R, Chow CY, Viray T, Johnson DS, Jiang Y, Patel SG, Ganly I, Schroeder CI, Lewis JS, King GF, Reiner T, Pillarsetty N. Na V1.7 targeted fluorescence imaging agents for nerve identification during intraoperative procedures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.06.588368. [PMID: 38617358 PMCID: PMC11014580 DOI: 10.1101/2024.04.06.588368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Surgeries and trauma result in traumatic and iatrogenic nerve damage that can result in a debilitating condition that approximately affects 189 million individuals worldwide. The risk of nerve injury during oncologic surgery is increased due to tumors displacing normal nerve location, blood turbidity, and past surgical procedures, which complicate even an experienced surgeon's ability to precisely locate vital nerves. Unfortunately, there is a glaring absence of contrast agents to assist surgeons in safeguarding vital nerves. To address this unmet clinical need, we leveraged the abundant expression of the voltage-gated sodium channel 1.7 (NaV1.7) as an intraoperative marker to access peripheral nerves in vivo, and visualized nerves for surgical guidance using a fluorescently-tagged version of a potent NaV1.7-targeted peptide, Tsp1a, derived from a Peruvian tarantula. We characterized the expression of NaV1.7 in sensory and motor peripheral nerves across mouse, primate, and human specimens and demonstrated universal expression. We synthesized and characterized a total of 10 fluorescently labeled Tsp1a-peptide conjugates to delineate nerves. We tested the ability of these peptide-conjugates to specifically accumulate in mouse nerves with a high signal-to-noise ratio in vivo. Using the best-performing candidate, Tsp1a-IR800, we performed thyroidectomies in non-human primates and demonstrated successful demarcation of the recurrent laryngeal and vagus nerves, which are commonly subjected to irreversible damage. The ability of Tsp1a to enhance nerve contrast during surgery provides opportunities to minimize nerve damage and revolutionize standards of care across various surgical specialties.
Collapse
Affiliation(s)
- Junior Gonzales
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA
| | - Dauren Adilbay
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA
| | - Paula Demetrio de Souza Franca
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA
- Department of Otorhinolaryngology and Head and Neck Surgery, Federal University of São Paulo, SP, Brazil
| | - Raik Artschwager
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA
| | - Chun Yuen Chow
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Research, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Tara Viray
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA
| | - Delissa S. Johnson
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA
| | - Yan Jiang
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Snehal G. Patel
- Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA
- Department of Otorhinolaryngology, Weill Cornell Medical College, 1300 York Avenue, New York, New York, 10065, USA
| | - Ian Ganly
- Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA
- Department of Otorhinolaryngology, Weill Cornell Medical College, 1300 York Avenue, New York, New York, 10065, USA
| | - Christina I. Schroeder
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Jason S. Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA
- Department of Pharmacology, Weill-Cornell Medical College, New York, New York, 10065, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York, 10065, USA
- Department of Radiology, Weill Cornell Medical College, 1300 York Avenue, New York, New York, 10065, USA
| | - Glenn F. King
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Research, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA
- Department of Pharmacology, Weill-Cornell Medical College, New York, New York, 10065, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York, 10065, USA
- Department of Radiology, Weill Cornell Medical College, 1300 York Avenue, New York, New York, 10065, USA
| | - Nagavarakishore Pillarsetty
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York, 10065, USA
- Department of Pharmacology, Weill-Cornell Medical College, New York, New York, 10065, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York, 10065, USA
- Department of Radiology, Weill Cornell Medical College, 1300 York Avenue, New York, New York, 10065, USA
| |
Collapse
|
21
|
Teng M, Liang X, Liu H, Li Z, Gao X, Zhang C, Cheng H, Chen H, Liu G. Cerenkov radiation shining a light for cancer theranostics. NANO TODAY 2024; 55:102174. [DOI: 10.1016/j.nantod.2024.102174] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
|
22
|
Si M, Lv L, Shi Y, Li Z, Zhai W, Luo X, Zhang L, Qian Y. Activatable Dual-Optical Molecular Probe for Bioimaging Superoxide Anion in Epilepsy. Anal Chem 2024; 96:4632-4638. [PMID: 38457631 DOI: 10.1021/acs.analchem.3c05641] [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: 03/10/2024]
Abstract
Superoxide anion (O2•-) plays a pivotal role in the generation of other reactive oxygen species within the body and is closely linked to epilepsy. Despite this connection, achieving precise imaging of O2•- during epilepsy pathology remains a formidable challenge. Herein, we develop an activatable molecular probe, CL-SA, to track the fluctuation of the level of O2•- in epilepsy through simultaneous fluorescence imaging and chemiluminescence sensing. The developed probe CL-SA demonstrated its efficacy in imaging of O2•- in neuronal cells, showcasing its dual optical imaging capability for O2•- in vitro. Furthermore, CL-SA was successfully used to observe aberrantly expressed O2•- in a mouse model of epilepsy. Overall, CL-SA provides us with a valuable tool for chemical and biomedical studies of O2•-, promoting the investigation of O2•- fluctuations in epilepsy, as well as providing a reliable means to explore the diagnosis and therapy of epilepsy.
Collapse
Affiliation(s)
- Mingran Si
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210046, China
| | - Li Lv
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Public Experimental Research Center, Xuzhou Medical University, Xuzhou 221002, China
| | - Yifan Shi
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Public Experimental Research Center, Xuzhou Medical University, Xuzhou 221002, China
| | - Zheng Li
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210046, China
| | - Wenjing Zhai
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Public Experimental Research Center, Xuzhou Medical University, Xuzhou 221002, China
| | - Xiangjie Luo
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210046, China
| | - Ling Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Public Experimental Research Center, Xuzhou Medical University, Xuzhou 221002, China
| | - Yong Qian
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210046, China
| |
Collapse
|
23
|
Giuliani S, Paraboschi I, McNair A, Smith M, Rankin KS, Elson DS, Paleri V, Leff D, Stasiuk G, Anderson J. Monoclonal Antibodies for Targeted Fluorescence-Guided Surgery: A Review of Applicability across Multiple Solid Tumors. Cancers (Basel) 2024; 16:1045. [PMID: 38473402 PMCID: PMC10931077 DOI: 10.3390/cancers16051045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 02/25/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
This study aims to review the status of the clinical use of monoclonal antibodies (mAbs) that have completed or are in ongoing clinical trials for targeted fluorescence-guided surgery (T-FGS) for the intraoperative identification of the tumor margins of extra-hematological solid tumors. For each of them, the targeted antigen, the mAb generic/commercial name and format, and clinical indications are presented, together with utility, doses, and the timing of administration. Based on the current scientific evidence in humans, the top three mAbs that could be prepared in a GMP-compliant bank ready to be delivered for surgical purposes are proposed to speed up the translation to the operating room and produce a few readily available "off-the-shelf" injectable fluorescent probes for safer and more effective solid tumor resection.
Collapse
Affiliation(s)
- Stefano Giuliani
- Wellcome/EPSRC Centre for Interventional and Surgical Sciences, University College London, London W1W 7TY, UK
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK;
- Department of Specialist Neonatal and Paediatric Surgery, Great Ormond Street Hospital for Children NHS Trust, London WC1N 3JH, UK
| | - Irene Paraboschi
- Department of Biomedical and Clinical Science, University of Milano, 20157 Milan, Italy;
| | - Angus McNair
- National Institute for Health Research Bristol Biomedical Research Centre, Bristol Centre for Surgical Research, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2PS, UK;
- Department of Gastrointestinal Surgery, North Bristol NHS Trust, Bristol BS10 5NB, UK
| | - Myles Smith
- The Sarcoma, Melanoma and Rare Tumours Unit, The Royal Marsden Hospital, Institute Cancer of Research, London SW3 6JJ, UK;
| | - Kenneth S. Rankin
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
- North of England Bone and Soft Tissue Tumour Service, Royal Victoria Infirmary, Newcastle upon Tyne NE1 4LP, UK
| | - Daniel S. Elson
- Hamlyn Centre for Robotic Surgery, Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK;
| | - Vinidh Paleri
- Head and Neck Unit, The Royal Marsden Hospitals, London SW3 6JJ, UK;
| | - Daniel Leff
- Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK;
| | - Graeme Stasiuk
- Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King’s College London, St Thomas’ Hospital, London SE1 7EH, UK;
| | - John Anderson
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK;
| |
Collapse
|
24
|
Ndabakuranye JP, Belcourt J, Sharma D, O'Connell CD, Mondal V, Srivastava SK, Stacey A, Long S, Fleiss B, Ahnood A. Miniature fluorescence sensor for quantitative detection of brain tumour. LAB ON A CHIP 2024; 24:946-954. [PMID: 38275166 DOI: 10.1039/d3lc00982c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
Fluorescence-guided surgery has emerged as a vital tool for tumour resection procedures. As well as intraoperative tumour visualisation, 5-ALA-induced PpIX provides an avenue for quantitative tumour identification based on ratiometric fluorescence measurement. To this end, fluorescence imaging and fibre-based probes have enabled more precise demarcation between the cancerous and healthy tissues. These sensing approaches, which rely on collecting the fluorescence light from the tumour resection site and its "remote" spectral sensing, introduce challenges associated with optical losses. In this work, we demonstrate the viability of tumour detection at the resection site using a miniature fluorescence measurement system. Unlike the current bulky systems, which necessitate remote measurement, we have adopted a millimetre-sized spectral sensor chip for quantitative fluorescence measurements. A reliable measurement at the resection site requires a stable optical window between the tissue and the optoelectronic system. This is achieved using an antifouling diamond window, which provides stable optical transparency. The system achieved a sensitivity of 92.3% and specificity of 98.3% in detecting a surrogate tumour at a resolution of 1 × 1 mm2. As well as addressing losses associated with collecting and coupling fluorescence light in the current 'remote' sensing approaches, the small size of the system introduced in this work paves the way for its direct integration with the tumour resection tools with the aim of more accurate interoperative tumour identification.
Collapse
Affiliation(s)
| | | | - Deepak Sharma
- School of Engineering, RMIT University, VIC 3000, Australia.
- Photovoltaic Metrology Section, Advanced Materials and Device Metrology Division, CSIR-National Physical Laboratory, New Delhi, 110012, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Cathal D O'Connell
- School of Engineering, RMIT University, VIC 3000, Australia.
- Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, VIC 3065, Australia
| | - Victor Mondal
- School of Health and Biomedical Sciences, RMIT University, VIC 3000, Australia
| | - Sanjay K Srivastava
- Photovoltaic Metrology Section, Advanced Materials and Device Metrology Division, CSIR-National Physical Laboratory, New Delhi, 110012, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Alastair Stacey
- School of Science, RMIT University, VIC 3000, Australia
- Princeton Plasma Physics Laboratory, Princeton University, Princeton, 08540 New Jersey, USA
| | - Sam Long
- Veterinary Referral Hospital, Victoria, Australia
| | - Bobbi Fleiss
- School of Health and Biomedical Sciences, RMIT University, VIC 3000, Australia
| | - Arman Ahnood
- School of Engineering, RMIT University, VIC 3000, Australia.
| |
Collapse
|
25
|
Zhou X, Fan Y, Li S, Zhang K, Pei Y, Zeng Y, Kang X, Zhao L, Chen H, Qin Y, Feng W, Liu L, Wu L. Molecular Engineering of Bright NIR-I/NIR-II Nanofluorophores for High-Resolution Bioimaging and Tumor Detection in Vivo. NANO LETTERS 2024; 24:1792-1800. [PMID: 38278136 DOI: 10.1021/acs.nanolett.3c04976] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2024]
Abstract
A comprehensive approach for the construction of NIR-I/NIR-II nanofluorophores with exceptional brightness and excellent chemo- and photostability has been developed. This study first confirmed that the amphiphilic molecules with stronger hydrophobic moieties and weaker hydrophilic moieties are superior candidates for constructing brighter nanofluorophores, which are attributed to its higher efficiency in suppressing the intramolecular charge transfer/aggregation-caused fluorescence quenching of donor-acceptor-donor type fluorophores. The prepared nanofluorophore demonstrates a fluorescence quantum yield exceeding 4.5% in aqueous solution and exhibits a strong NIR-II tail emission up to 1300 nm. The superior performance of the nanofluorophore enabled the achievement of high-resolution whole-body vessel imaging and brain vessel imaging, as well as high-contrast fluorescence imaging of the lymphatic system in vivo. Furthermore, their potential for highly sensitive fluorescence detection of tiny tumors in vivo has been successfully confirmed, thus supporting their future applications in precise fluorescence imaging-guided surgery in the early stages of cancer.
Collapse
Affiliation(s)
- Xiaobo Zhou
- School of Public Health, Nantong Key Laboratory of Public Health and Medical Analysis, Nantong University, Nantong 226019, Jiangsu, China
| | - Yiwei Fan
- Department of Chemistry & State Key Laboratory of Molecular Engineering of Polymers & Academy for Engineering and Technology, Fudan University, Shanghai 200433, China
| | - Shijie Li
- School of Public Health, Nantong Key Laboratory of Public Health and Medical Analysis, Nantong University, Nantong 226019, Jiangsu, China
| | - Ke Zhang
- School of Public Health, Nantong Key Laboratory of Public Health and Medical Analysis, Nantong University, Nantong 226019, Jiangsu, China
| | - Yuetian Pei
- Department of Chemistry & State Key Laboratory of Molecular Engineering of Polymers & Academy for Engineering and Technology, Fudan University, Shanghai 200433, China
| | - Yuhan Zeng
- School of Public Health, Nantong Key Laboratory of Public Health and Medical Analysis, Nantong University, Nantong 226019, Jiangsu, China
| | - Xiaoxia Kang
- School of Public Health, Nantong Key Laboratory of Public Health and Medical Analysis, Nantong University, Nantong 226019, Jiangsu, China
| | - Lingfeng Zhao
- School of Public Health, Nantong Key Laboratory of Public Health and Medical Analysis, Nantong University, Nantong 226019, Jiangsu, China
| | - Hao Chen
- Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yuling Qin
- School of Public Health, Nantong Key Laboratory of Public Health and Medical Analysis, Nantong University, Nantong 226019, Jiangsu, China
| | - Wei Feng
- Department of Chemistry & State Key Laboratory of Molecular Engineering of Polymers & Academy for Engineering and Technology, Fudan University, Shanghai 200433, China
| | - Lingxiao Liu
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Li Wu
- School of Public Health, Nantong Key Laboratory of Public Health and Medical Analysis, Nantong University, Nantong 226019, Jiangsu, China
| |
Collapse
|
26
|
Filip P, Lerner DK, Kominsky E, Schupper A, Liu K, Khan NM, Roof S, Hadjipanayis C, Genden E, Iloreta AMC. 5-Aminolevulinic Acid Fluorescence-Guided Surgery in Head and Neck Squamous Cell Carcinoma. Laryngoscope 2024; 134:741-748. [PMID: 37540051 DOI: 10.1002/lary.30910] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 08/05/2023]
Abstract
OBJECTIVES To determine the utility of 5-aminolevulinic acid (5-ALA) fluorescence for resection of head and neck carcinoma. METHODS In this prospective pilot trial, 5-ALA was administered as an oral suspension 3-5 h prior to induction of anesthesia for resection of head and neck squamous cell carcinoma (HNSCC). Following resection, 405 nm blue light was applied, and fluorescence of the tumor as well as the surgical bed was recorded. Specimen fluorescence intensity was graded categorically as none (score = 0), mild (1), moderate (2), or robust (3) by the operating surgeon intraoperatively and corroborated with final pathologic diagnosis. RESULTS Seven patients underwent resection with 5-ALA. Five (83%) were male with an age range of 33-82 years (mean = 60). Sites included nasal cavity (n = 3), oral cavity (n = 3), and the larynx (n = 1). All specimens demonstrated robust fluorescence when 5-ALA was administered 3-5 h preoperatively. 5-ALA fluorescence predicted the presence of perineural invasion, a positive margin, and metastatic lymphadenopathy. Two patients had acute photosensitivity reactions, and one patient had a temporary elevation of hepatic enzymes. CONCLUSIONS 5-ALA induces robust intraoperative fluorescence of HNSCC, capable of demonstrating a positive margin, perineural invasion, and metastatic nodal disease. Although no conclusions are there about the safety of this drug in the head and neck cancer population, our study parallels the extensive safety data in the neurosurgical literature. Future applications may include intraoperative assessment of margin status, diagnostic accuracy, and impacts on survival. LEVEL OF EVIDENCE 4 Laryngoscope, 134:741-748, 2024.
Collapse
Affiliation(s)
- Peter Filip
- Department of Otolaryngology-Head and Neck Surgery, Icahn School of Medicine at Mount Sinai, New York City, New York, U.S.A
| | - David K Lerner
- Department of Otolaryngology-Head and Neck Surgery, Icahn School of Medicine at Mount Sinai, New York City, New York, U.S.A
| | - Evan Kominsky
- Department of Otolaryngology-Head and Neck Surgery, Icahn School of Medicine at Mount Sinai, New York City, New York, U.S.A
| | - Alexander Schupper
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York City, New York, U.S.A
| | - Katherine Liu
- Department of Otolaryngology-Head and Neck Surgery, Icahn School of Medicine at Mount Sinai, New York City, New York, U.S.A
| | - Nazir Mohemmed Khan
- Department of Otolaryngology-Head and Neck Surgery, Icahn School of Medicine at Mount Sinai, New York City, New York, U.S.A
| | - Scott Roof
- Department of Otolaryngology-Head and Neck Surgery, Icahn School of Medicine at Mount Sinai, New York City, New York, U.S.A
| | | | - Eric Genden
- Department of Otolaryngology-Head and Neck Surgery, Icahn School of Medicine at Mount Sinai, New York City, New York, U.S.A
| | - Alfred M C Iloreta
- Department of Otolaryngology-Head and Neck Surgery, Icahn School of Medicine at Mount Sinai, New York City, New York, U.S.A
| |
Collapse
|
27
|
Yang Y, Jiang Q, Zhang F. Nanocrystals for Deep-Tissue In Vivo Luminescence Imaging in the Near-Infrared Region. Chem Rev 2024; 124:554-628. [PMID: 37991799 DOI: 10.1021/acs.chemrev.3c00506] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
In vivo imaging technologies have emerged as a powerful tool for both fundamental research and clinical practice. In particular, luminescence imaging in the tissue-transparent near-infrared (NIR, 700-1700 nm) region offers tremendous potential for visualizing biological architectures and pathophysiological events in living subjects with deep tissue penetration and high imaging contrast owing to the reduced light-tissue interactions of absorption, scattering, and autofluorescence. The distinctive quantum effects of nanocrystals have been harnessed to achieve exceptional photophysical properties, establishing them as a promising category of luminescent probes. In this comprehensive review, the interactions between light and biological tissues, as well as the advantages of NIR light for in vivo luminescence imaging, are initially elaborated. Subsequently, we focus on achieving deep tissue penetration and improved imaging contrast by optimizing the performance of nanocrystal fluorophores. The ingenious design strategies of NIR nanocrystal probes are discussed, along with their respective biomedical applications in versatile in vivo luminescence imaging modalities. Finally, thought-provoking reflections on the challenges and prospects for future clinical translation of nanocrystal-based in vivo luminescence imaging in the NIR region are wisely provided.
Collapse
Affiliation(s)
- Yang Yang
- College of Energy Materials and Chemistry, State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010021, China
| | - Qunying Jiang
- College of Energy Materials and Chemistry, State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010021, China
| | - Fan Zhang
- College of Energy Materials and Chemistry, State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010021, China
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, China
| |
Collapse
|
28
|
Yang Z, Zhang Y, Tang L, Yang X, Song L, Shen C, Zvyagin AV, Li Y, Yang B, Lin Q. "All in one" nanoprobe Au-TTF-1 for target FL/CT bioimaging, machine learning technology and imaging-guided photothermal therapy against lung adenocarcinoma. J Nanobiotechnology 2024; 22:22. [PMID: 38184620 PMCID: PMC10770976 DOI: 10.1186/s12951-023-02280-9] [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: 11/04/2023] [Accepted: 12/19/2023] [Indexed: 01/08/2024] Open
Abstract
The accurate preoperative diagnosis and tracking of lung adenocarcinoma is hindered by non-targeting and diffusion of dyes used for marking tumors. Hence, there is an urgent need to develop a practical nanoprobe for tracing lung adenocarcinoma precisely even treating them noninvasively. Herein, Gold nanoclusters (AuNCs) conjugate with thyroid transcription factor-1 (TTF-1) antibody, then multifunctional nanoprobe Au-TTF-1 is designed and synthesized, which underscores the paramount importance of advancing the machine learning diagnosis and bioimaging-guided treatment of lung adenocarcinoma. Bright fluorescence (FL) and strong CT signal of Au-TTF-1 set the stage for tracking. Furthermore, the high specificity of TTF-1 antibody facilitates selective targeting of lung adenocarcinoma cells as compared to common lung epithelial cells, so machine learning software Lung adenocarcinoma auxiliary detection system was designed, which combined with Au-TTF-1 to assist the intelligent recognition of lung adenocarcinoma jointly. Besides, Au-TTF-1 not only contributes to intuitive and targeted visualization, but also guides the following noninvasive photothermal treatment. The boundaries of tumor are light up by Au-TTF-1 for navigation, it penetrates into tumor and implements noninvasive photothermal treatment, resulting in ablating tumors in vivo locally. Above all, Au-TTF-1 serves as a key platform for target bio-imaging navigation, machine learning diagnosis and synergistic PTT as a single nanoprobe, which demonstrates attractive performance on lung adenocarcinoma.
Collapse
Affiliation(s)
- Zhe Yang
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Yujia Zhang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, 130021, China
| | - Lu Tang
- Department of Breast, China-Japan Union Hospital of Jilin University, Changchun, 130031, China
| | - Xiao Yang
- College of Computer Science and Technology Jilin University, Changchun, 130012, China
| | - Lei Song
- Department of Breast, China-Japan Union Hospital of Jilin University, Changchun, 130031, China
| | - Chun Shen
- College of Computer Science and Technology Jilin University, Changchun, 130012, China
| | - Andrei V Zvyagin
- Australian Research Council Centre of Excellence for Nanoscale Biophotonics, Macquarie University, Sydney, NSW, 2109, Australia
| | - Yang Li
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, 130021, China.
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun, 130012, China
| | - Quan Lin
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun, 130012, China.
| |
Collapse
|
29
|
Yuan L, Su Y, Zhang R, Gao J, Yu B, Cong H, Shen Y. NIR-II organic small molecule probe for labeling lymph nodes and guiding tumor imaging. Talanta 2024; 266:125123. [PMID: 37639868 DOI: 10.1016/j.talanta.2023.125123] [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: 06/15/2023] [Revised: 08/20/2023] [Accepted: 08/24/2023] [Indexed: 08/31/2023]
Abstract
Organic small molecule fluorescent groups have injected new material support into the field of medical imaging due to their unique luminescence mechanism and easy tuning of structure. The great potential of NIR-II window imaging forces us to continuously optimize the structure of organic fluorophores to design better fluorescent molecules for fluorescence imaging-guided surgery. An ideal organic small molecule fluorescent group: it can penetrate into the inside of the organism, clearly present the internal structure and the edge contour of different tissues, so as to perfectly achieve internal imaging and accurately guide external surgery. In vivo, fluorescent groups do not damage normal tissues and organs. However, problems such as low quantum yield and poor biocompatibility greatly limit the clinical transformation of NIR-II fluorescent small molecules. To avoid the shortcomings of NIR-II fluorescent probes as much as possible and better realize image-guided surgery, in this experiment, the biplane donor unit was incorporated into the twisted D-π-A-π-D structure to expand the conjugated structure of the fluorescent group, which not only realized NIR-II emission, but also had high quantum yield and biosafety.
Collapse
Affiliation(s)
- Lin Yuan
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Yingbin Su
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Runfeng Zhang
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Jie Gao
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Bing Yu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, 266071, China.
| | - Hailin Cong
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, 266071, China; School of Materials Science and Engineering, Shandong University of Technology, Zibo, 255000, China.
| | - Youqing Shen
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China; Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, And Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| |
Collapse
|
30
|
Petusseau AF, Streeter SS, Ulku A, Feng Y, Samkoe KS, Bruschini C, Charbon E, Pogue BW, Bruza P. Subsurface fluorescence time-of-flight imaging using a large-format single-photon avalanche diode sensor for tumor depth assessment. JOURNAL OF BIOMEDICAL OPTICS 2024; 29:016004. [PMID: 38235320 PMCID: PMC10794045 DOI: 10.1117/1.jbo.29.1.016004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 12/06/2023] [Accepted: 12/20/2023] [Indexed: 01/19/2024]
Abstract
Significance Fluorescence guidance is used clinically by surgeons to visualize anatomical and/or physiological phenomena in the surgical field that are difficult or impossible to detect by the naked eye. Such phenomena include tissue perfusion or molecular phenotypic information about the disease being resected. Conventional fluorescence-guided surgery relies on long, microsecond scale laser pulses to excite fluorescent probes. However, this technique only provides two-dimensional information; crucial depth information, such as the location of malignancy below the tissue surface, is not provided. Aim We developed a depth sensing imaging technique using light detection and ranging (LiDAR) time-of-flight (TOF) technology to sense the depth of target tissue while overcoming the influence of tissue optical properties and fluorescent probe concentration. Approach The technology is based on a large-format (512 × 512 pixel ), binary, gated, single-photon avalanche diode (SPAD) sensor with an 18 ps time-gate step, synchronized with a picosecond pulsed laser. The fast response of the sensor was developed and tested for its ability to quantify fluorescent inclusions at depth and optical properties in tissue-like phantoms through analytical model fitting of the fast temporal remission data. Results After calibration and algorithmic extraction of the data, the SPAD LiDAR technique allowed for sub-mm resolution depth sensing of fluorescent inclusions embedded in tissue-like phantoms, up to a maximum of 5 mm in depth. The approach provides robust depth sensing even in the presence of variable tissue optical properties and separates the effects of fluorescence depth from absorption and scattering variations. Conclusions LiDAR TOF fluorescence imaging using an SPAD camera provides both fluorescence intensity images and the temporal profile of fluorescence, which can be used to determine the depth at which the signal is emitted over a wide field of view. The proposed tool enables fluorescence imaging at a higher depth in tissue and with higher spatial precision than standard, steady-state fluorescence imaging tools, such as intensity-based near-infrared fluorescence imaging, optical coherence tomography, Raman spectroscopy, or confocal microscopy. Integration of this technique into a standard surgical tool could enable rapid, more accurate estimation of resection boundaries, thereby improving the surgeon's efficacy and efficiency, and ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Arthur F. Petusseau
- Dartmouth College, Thayer School of Engineering and Dartmouth Cancer Center, Hanover, New Hampshire, United States
| | - Samuel S. Streeter
- Geisel School of Medicine at Dartmouth, Department of Orthopaedics, Hanover, New Hampshire, United States
| | - Arin Ulku
- Ecole polytechnique fédérale de Lausanne, Advanced Quantum Architecture Laboratory, Neuchâtel, Switzerland
| | - Yichen Feng
- Geisel School of Medicine at Dartmouth, Department of Surgery, Hanover, New Hampshire, United States
| | - Kimberley S. Samkoe
- Geisel School of Medicine at Dartmouth, Department of Surgery, Hanover, New Hampshire, United States
| | - Claudio Bruschini
- Ecole polytechnique fédérale de Lausanne, Advanced Quantum Architecture Laboratory, Neuchâtel, Switzerland
| | - Edoardo Charbon
- Ecole polytechnique fédérale de Lausanne, Advanced Quantum Architecture Laboratory, Neuchâtel, Switzerland
| | - Brian W. Pogue
- Dartmouth College, Thayer School of Engineering and Dartmouth Cancer Center, Hanover, New Hampshire, United States
- University of Wisconsin–Madison, Department of Medical Physics, Madison, Wisconsin, United States
| | - Petr Bruza
- Dartmouth College, Thayer School of Engineering and Dartmouth Cancer Center, Hanover, New Hampshire, United States
| |
Collapse
|
31
|
Xu Z, Tao W, Qian J, Zhao H, Peng Y, Sun T, Gao G, Ling C, Li P, Chen J, Ling Y. Dual Tumor-Selective β-Carboline-Based Fluorescent Probe for High-Contrast/Rapid Diagnosis of Clinical Tumor Tissues. Mol Pharm 2024; 21:152-163. [PMID: 38113058 DOI: 10.1021/acs.molpharmaceut.3c00689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Given that precise/rapid intraoperative tumor margin identification is still challenging, novel fluorescent probes HY and HYM, based on acidic tumor microenvironment (TME) activation and organic anion transporting polypeptide (OATPs)-mediated selective uptake, were constructed and synthesized. Both of them possessed acidic pH-activatable and reversible fluorescence as well as large Stokes shift. Compared with HY, HYM had a higher (over 9-fold) enhancement in fluorescence with pH ranging from 7.6 to 4.0, and the fluorescence quantum yield of HYM (ΦF = 0.49) at pH = 4.0 was 8-fold stronger than that (ΦF = 0.06) at pH = 7.4. Mechanism research demonstrated that acidic TME-induced protonation of the pyridine N atom on β-carbolines accounted for the pH-sensitive fluorescence by influencing the intramolecular charge transfer (ICT) effect. Furthermore, HYM selectively lit up cancer cells and tumor tissues not only by "off-on" fluorescence but also by OATPs (overexpressed on cancer cells)-mediated cancer cellular internalization, offering dual tumor selectivity for precise visualization of tumor mass and intraoperative guidance upon in situ spraying. Most importantly, HYM enabled rapid and high-contrast (tumor-to-normal tissue ratios > 6) human tumor margin identification in clinical tumor tissues by simple spraying within 6 min, being promising for aiding in clinical surgical resection.
Collapse
Affiliation(s)
- Zhongyuan Xu
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, 226001 Nantong, Jiangsu, P. R. China
- Department of Hepatobiliary Surgery, Nantong Third People's Hospital and the Third Affiliated Hospital of Nantong University, 226001 Nantong, Jiangsu, P. R. China
| | - Weizhi Tao
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, 226001 Nantong, Jiangsu, P. R. China
| | - Jianqiang Qian
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, 226001 Nantong, Jiangsu, P. R. China
| | - Huimin Zhao
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, 226001 Nantong, Jiangsu, P. R. China
| | - Yiqian Peng
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, 226001 Nantong, Jiangsu, P. R. China
| | - Tiantian Sun
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, 226001 Nantong, Jiangsu, P. R. China
| | - Ge Gao
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, 226001 Nantong, Jiangsu, P. R. China
| | - Changchun Ling
- Department of General Surgery, Affiliated Hospital of Nantong University, 226001 Nantong, Jiangsu, P. R. China
| | - Peng Li
- Department of General Surgery, Affiliated Hospital of Nantong University, 226001 Nantong, Jiangsu, P. R. China
| | - Jun Chen
- Department of Hepatobiliary Surgery, Nantong Third People's Hospital and the Third Affiliated Hospital of Nantong University, 226001 Nantong, Jiangsu, P. R. China
| | - Yong Ling
- School of Pharmacy and Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong Key Laboratory of Small Molecular Drug Innovation, Nantong University, 226001 Nantong, Jiangsu, P. R. China
| |
Collapse
|
32
|
Wang B, Tang C, Lin E, Jia X, Xie G, Li P, Li D, Yang Q, Guo X, Cao C, Shi X, Zou B, Cai C, Tian J, Hu Z, Li J. NIR-II fluorescence-guided liver cancer surgery by a small molecular HDAC6 targeting probe. EBioMedicine 2023; 98:104880. [PMID: 38035463 PMCID: PMC10698675 DOI: 10.1016/j.ebiom.2023.104880] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 11/05/2023] [Accepted: 11/05/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the sixth most common malignancy globally and ranks third in terms of both mortality and incidence rates. Surgical resection holds potential as a curative approach for HCC. However, the residual disease contributes to a high 5-year recurrence rate of 70%. Due to their excellent specificity and optical properties, fluorescence-targeted probes are deemed effective auxiliary tools for addressing residual lesions, enabling precise surgical diagnosis and treatment. Research indicates histone deacetylase 6 (HDAC6) overexpression in HCC cells, making it a potential imaging biomarker. This study designed a targeted small-molecule fluorescent probe, SeCF3-IRDye800cw (SeCF3-IRD800), operating within the Second near-infrared window (NIR-II, 1000-1700 nm). The study confirms the biocompatibility of SeCF3-IRD800 and proceeds to demonstrate its applications in imaging in vivo, fluorescence-guided surgery (FGS) for liver cancer, liver fibrosis imaging, and clinical samples incubation, thereby preliminarily validating its utility in liver cancer. METHODS SeCF3-IRD800 was synthesized by combining the near-infrared fluorescent dye IRDye800cw-NHS with an improved HDAC6 inhibitor. Initially, a HepG2-Luc subcutaneous tumor model (n = 12) was constructed to investigate the metabolic differences between SeCF3-IRD800 and ICG in vivo. Subsequently, HepG2-Luc (n = 12) and HCCLM3-Luc (n = 6) subcutaneous xenograft mouse models were used to assess in vivo targeting by SeCF3-IRD800. The HepG2-Luc orthotopic liver cancer model (n = 6) was employed to showcase the application of SeCF3-IRD800 in FGS. Liver fibrosis (n = 6) and HepG2-Luc orthotopic (n = 6) model imaging results were used to evaluate the impact of different pathological backgrounds on SeCF3-IRD800 imaging. Three groups of fresh HCC and normal liver samples from patients with liver cancer were utilized for SeCF3-IRD800 incubation ex vivo, while preclinical experiments illustrated its potential for clinical application. FINDINGS The HDAC6 inhibitor 6 (SeCF3) modified with trifluoromethyl was labeled with IRDy800CW-NHS to synthesize the small-molecule targeted probe SeCF3-IRD800, with NIR-II fluorescence signals. SeCF3-IRD800 was rapidly metabolized by the kidneys and exhibited excellent biocompatibility. In vivo validation demonstrated that SeCF3-IRD800 achieved optimal imaging within 8 h, displaying high tumor fluorescence intensity (7658.41 ± 933.34) and high tumor-to-background ratio (5.20 ± 1.04). Imaging experiments with various expression levels revealed its capacity for HDAC6-specific targeting across multiple HCC tumor models, suitable for NIR-II intraoperative imaging. Fluorescence-guided surgery experiments were found feasible and capable of detecting sub-visible 2 mm tumor lesions under white light, aiding surgical decision-making. Further imaging of liver fibrosis mice showed that SeCF3-IRD800's imaging efficacy remained unaffected by liver pathological conditions. Correlations were observed between HDAC6 expression levels and corresponding fluorescence intensity (R2 = 0.8124) among normal liver, liver fibrosis, and HCC tissues. SeCF3-IRD800 identified HDAC6-positive samples from patients with HCC, holding advantages for perspective intraoperative identification in liver cancer. Thus, the rapidly metabolized HDAC6-targeted small-molecule NIR-II fluorescence probe SeCF3-IRD800 holds significant clinical translational value. INTERPRETATION The successful application of NIR-II fluorescence-guided surgery in liver cancer indicates that SeCF3-IRD800 has great potential to improve the clinical diagnosis and treatment of liver cancer, and could be used as an auxiliary tool for surgical treatment of liver cancer without being affected by liver pathology. FUNDING This paper is supported by the National Natural Science Foundation of China (NSFC) (92,059,207, 62,027,901, 81,930,053, 81,227,901, 82,272,105, U21A20386 and 81,971,773), CAS Youth Interdisciplinary Team (JCTD-2021-08), the Zhuhai High-level Health Personnel Team Project (Zhuhai HLHPTP201703), and Guangdong Basic and Applied Basic Research Foundation under Grant No. 2022A1515011244.
Collapse
Affiliation(s)
- Bo Wang
- Department of Hepatobiliary Surgery and Liver Transplantation, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China; CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Chu Tang
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, 710071, China
| | - En Lin
- Department of Hepatobiliary Surgery and Liver Transplantation, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China; CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China
| | - Xiaohua Jia
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ganyuan Xie
- Department of Hepatobiliary Surgery and Liver Transplantation, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Peiping Li
- Department of Hepatobiliary Surgery and Liver Transplantation, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Decheng Li
- Department of Hepatobiliary Surgery and Liver Transplantation, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Qiyue Yang
- Key Laboratory of Digital Hepatobiliary Surgery, PLA, Institute of Hepatobiliary Surgery of Chinese PLA, Beijing, 100048, China
| | - Xiaoyong Guo
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China; Clinical College of Armed Police General Hospital of Anhui Medical University, Department of Gastroenterology of The Third Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Caiguang Cao
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaojing Shi
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Baojia Zou
- Department of Hepatobiliary Surgery and Liver Transplantation, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Chaonong Cai
- Department of Hepatobiliary Surgery and Liver Transplantation, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China; Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, 100191, China; Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, 710071, China.
| | - Zhenhua Hu
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China; School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Jian Li
- Department of Hepatobiliary Surgery and Liver Transplantation, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China.
| |
Collapse
|
33
|
Imanishi H, Nishimura T, Shimojo Y, Awazu K. Deep learning based depth map estimation of protoporphyrin IX in turbid media using dual wavelength excitation fluorescence. BIOMEDICAL OPTICS EXPRESS 2023; 14:5254-5266. [PMID: 37854564 PMCID: PMC10581804 DOI: 10.1364/boe.500022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/09/2023] [Accepted: 09/11/2023] [Indexed: 10/20/2023]
Abstract
This study presents a depth map estimation of fluorescent objects in turbid media, such as biological tissue based on fluorescence observation by two-wavelength excitation and deep learning-based processing. A U-Net-based convolutional neural network is adapted for fluorophore depth maps from the ratiometric information of the two-wavelength excitation fluorescence. The proposed method offers depth map estimation from wide-field fluorescence images with rapid processing. The feasibility of the proposed method was demonstrated experimentally by estimating the depth map of protoporphyrin IX, a recognized cancer biomarker, at different depths within an optical phantom.
Collapse
Affiliation(s)
- Hinano Imanishi
- Graduate School of Engineering, Osaka University, Yamadaoka 2-1, Suita, Osaka 565-0871, Japan
| | - Takahiro Nishimura
- Graduate School of Engineering, Osaka University, Yamadaoka 2-1, Suita, Osaka 565-0871, Japan
| | - Yu Shimojo
- Graduate School of Engineering, Osaka University, Yamadaoka 2-1, Suita, Osaka 565-0871, Japan
- Graduate School of Medicine, Osaka Metropolitan University, Asahimachi 1-4-3, Abeno-ku, Osaka 545-8585, Japan
- Research Fellow of Japan Society for the Promotion of Science, Kojimachi 5-3-1, Chiyoda-ku, Tokyo 102-0083, Japan
| | - Kunio Awazu
- Graduate School of Engineering, Osaka University, Yamadaoka 2-1, Suita, Osaka 565-0871, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| |
Collapse
|
34
|
Mirbeik A, Ebadi N. Deep learning for tumor margin identification in electromagnetic imaging. Sci Rep 2023; 13:15925. [PMID: 37741854 PMCID: PMC10517989 DOI: 10.1038/s41598-023-42625-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 09/12/2023] [Indexed: 09/25/2023] Open
Abstract
In this work, a novel method for tumor margin identification in electromagnetic imaging is proposed to optimize the tumor removal surgery. This capability will enable the visualization of the border of the cancerous tissue for the surgeon prior or during the excision surgery. To this end, the border between the normal and tumor parts needs to be identified. Therefore, the images need to be segmented into tumor and normal areas. We propose a deep learning technique which divides the electromagnetic images into two regions: tumor and normal, with high accuracy. We formulate deep learning from a perspective relevant to electromagnetic image reconstruction. A recurrent auto-encoder network architecture (termed here DeepTMI) is presented. The effectiveness of the algorithm is demonstrated by segmenting the reconstructed images of an experimental tissue-mimicking phantom. The structure similarity measure (SSIM) and mean-square-error (MSE) average of normalized reconstructed results by the DeepTMI method are about 0.94 and 0.04 respectively, while that average obtained from the conventional backpropagation (BP) method can hardly overcome 0.35 and 0.41 respectively.
Collapse
Affiliation(s)
- Amir Mirbeik
- RadioSight LLC, Hoboken, NJ, 07030, USA
- Department of Electrical and Computer Engineering, Stevens Institute of Technology, 1 Castle Point Ter, Hoboken, NJ, 07030, USA
| | - Negar Ebadi
- Department of Electrical and Computer Engineering, Stevens Institute of Technology, 1 Castle Point Ter, Hoboken, NJ, 07030, USA.
- Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
35
|
Mondal A, Kang J, Kim D. Recent Progress in Fluorescent Probes for Real-Time Monitoring of Glioblastoma. ACS APPLIED BIO MATERIALS 2023; 6:3484-3503. [PMID: 36917648 DOI: 10.1021/acsabm.3c00052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Treating glioblastoma (GBM) by resecting to a large extent can prolong a patient's survival by controlling the tumor cells, but excessive resection may produce postoperative complications by perturbing the brain structures. Therefore, various imaging procedures have been employed to successfully diagnose and resect with utmost caution and to protect vital structural or functional features. Fluorescence tagging is generally used as an intraoperative imaging technique in glioma cells in collaboration with other surgical tools such as MRI and navigation methods. However, the existing fluorescent probes may have several limitations, including poor selectivity, less photostability, false signals, and intraoperative re-administration when used in clinical and preclinical studies for glioma surgery. The involvement of smart fluorogenic materials, specifically fluorescent dyes, and biomarker-amended cell-penetrable fluorescent probes have noteworthy advantages for precise glioma imaging. This review outlines the contemporary advancements of fluorescent probes for imaging glioma cells along with their challenges and visions, with the anticipation to develop next-generation smart glioblastoma detection modalities.
Collapse
Affiliation(s)
- Amita Mondal
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jisoo Kang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, South Korea
| | - Dokyoung Kim
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, South Korea
- Center for Converging Humanities, Kyung Hee University, Seoul 02447, Republic of Korea
- Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Core Research Institute (CRI), Kyung Hee University, Seoul 02447, Republic of Korea
- Materials Research Science and Engineering Center, University of California at San Diego, 9500 Gilman Drive La Jolla, California 92093, United States
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
36
|
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
|
37
|
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
|
38
|
Bianconi A, Bonada M, Zeppa P, Colonna S, Tartara F, Melcarne A, Garbossa D, Cofano F. How Reliable Is Fluorescence-Guided Surgery in Low-Grade Gliomas? A Systematic Review Concerning Different Fluorophores. Cancers (Basel) 2023; 15:4130. [PMID: 37627158 PMCID: PMC10452554 DOI: 10.3390/cancers15164130] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Fluorescence-guided surgery has been increasingly used to support glioma surgery with the purpose of obtaining a maximal safe resection, in particular in high-grade gliomas, while its role is less definitely assessed in low-grade gliomas. METHODS A systematic review was conducted. 5-aminolevulinic acid, sodium fluorescein, indocyanine green and tozuleristide were taken into account. The main considered outcome was the fluorescence rate, defined as the number of patients in whom positive fluorescence was detected out of the total number of patients. Only low-grade gliomas were considered, and data were grouped according to single fluorophores. RESULTS 16 papers about 5-aminolevulinic acid, 4 about sodium fluorescein, 2 about indocyanine green and 1 about tozuleristide were included in the systematic review. Regarding 5-aminolevulinic acid, a total of 467 low-grade glioma patients were included, and fluorescence positivity was detected in 34 out of 451 Grade II tumors (7.3%); while in Grade I tumors, fluorescence positivity was detected in 9 out of 16 cases. In 16 sodium fluorescein patients, seven positive fluorescent cases were detected. As far as indocyanine is concerned, two studies accounting for six patients (three positive) were included, while for tozuleristide, a single clinical trial with eight patients (two positive) was retrieved. CONCLUSIONS The current evidence does not support the routine use of 5-aminolevulinic acid or sodium fluorescein with a standard operating microscope because of the low fluorescence rates. New molecules, including tozuleristide, and new techniques for fluorescence detection have shown promising results; however, their use still needs to be clinically validated on a large scale.
Collapse
Affiliation(s)
- Andrea Bianconi
- Neurosurgery, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.B.); (P.Z.); (A.M.); (D.G.); (F.C.)
| | - Marta Bonada
- Neurosurgery, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.B.); (P.Z.); (A.M.); (D.G.); (F.C.)
| | - Pietro Zeppa
- Neurosurgery, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.B.); (P.Z.); (A.M.); (D.G.); (F.C.)
| | - Stefano Colonna
- Neurosurgery, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.B.); (P.Z.); (A.M.); (D.G.); (F.C.)
| | - Fulvio Tartara
- Headache Science and Neurorehabilitation Center, IRCCS Mondino Foundation, Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| | - Antonio Melcarne
- Neurosurgery, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.B.); (P.Z.); (A.M.); (D.G.); (F.C.)
| | - Diego Garbossa
- Neurosurgery, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.B.); (P.Z.); (A.M.); (D.G.); (F.C.)
| | - Fabio Cofano
- Neurosurgery, Department of Neurosciences, University of Turin, 10126 Turin, Italy; (M.B.); (P.Z.); (A.M.); (D.G.); (F.C.)
- Humanitas Gradenigo, 10100 Turin, Italy
| |
Collapse
|
39
|
Zhang F, Xu J, Yue Y, Wang Y, Sun J, Song D, Zhang C, Qu L, Zhu S, Zhang J, Yang B. Three-dimensional histological electrophoresis enables fast automatic distinguishment of cancer margins and lymph node metastases. SCIENCE ADVANCES 2023; 9:eadg2690. [PMID: 37390200 PMCID: PMC10313175 DOI: 10.1126/sciadv.adg2690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 05/25/2023] [Indexed: 07/02/2023]
Abstract
Tissue diagnosis is important during surgical excision of solid tumors for margin evaluation. Conventional histopathologic methods rely heavily on image-based visual diagnosis by specialized pathologists, which can be time-consuming and subjective. We report a three-dimensional (3D) histological electrophoresis system for rapid labeling and separation of the proteins within tissue sections, providing a more precise assessment of tumor-positive margin in surgically resected tissues. The 3D histological electrophoresis system uses a tumor-seeking dye labeling strategy to visualize the distribution of tumor-specific proteins within sections and a tumor finder that automatically predicts the tumor contour. We successfully demonstrated the system's capability to predict the tumor contours from five murine xenograft models and distinguish the tumor-invaded region of sentinel lymph nodes. Specifically, we used the system to accurately assess tumor-positive margins from 14 patients with cancer. Our 3D histological electrophoresis system serves as an intraoperative tissue assessment technology for more accurate and automatic pathologic diagnosis.
Collapse
Affiliation(s)
- Feiran Zhang
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Jiajun Xu
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Ying Yue
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Yajun Wang
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Jianing Sun
- School of Mathematics and Statistics, Northeast Normal University, Changchun 130024, P. R. China
| | - Dong Song
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Chengbin Zhang
- Department of Pathology, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Limei Qu
- Department of Pathology, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Shoujun Zhu
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Junhu Zhang
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun 130021, P. R. China
| |
Collapse
|
40
|
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
|
41
|
Kim J, Kim H, Yoon YS, Kim CW, Hong SM, Kim S, Choi D, Chun J, Hong SW, Hwang SW, Park SH, Yang DH, Ye BD, Byeon JS, Yang SK, Kim SY, Myung SJ. Investigation of artificial intelligence integrated fluorescence endoscopy image analysis with indocyanine green for interpretation of precancerous lesions in colon cancer. PLoS One 2023; 18:e0286189. [PMID: 37228164 DOI: 10.1371/journal.pone.0286189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023] Open
Abstract
Indocyanine green (ICG) has been used in clinical practice for more than 40 years and its safety and preferential accumulation in tumors has been reported for various tumor types, including colon cancer. However, reports on clinical assessments of ICG-based molecular endoscopy imaging for precancerous lesions are scarce. We determined visualization ability of ICG fluorescence endoscopy in colitis-associated colon cancer using 30 lesions from an azoxymethane/dextran sulfate sodium (AOM/DSS) mouse model and 16 colon cancer patient tissue-samples. With a total of 60 images (optical, fluorescence) obtained during endoscopy observation of mouse colon cancer, we used deep learning network to predict four classes (Normal, Dysplasia, Adenoma, and Carcinoma) of colorectal cancer development. ICG could detect 100% of carcinoma, 90% of adenoma, and 57% of dysplasia, with little background signal at 30 min after injection via real-time fluorescence endoscopy. Correlation analysis with immunohistochemistry revealed a positive correlation of ICG with inducible nitric oxide synthase (iNOS; r > 0.5). Increased expression of iNOS resulted in increased levels of cellular nitric oxide in cancer cells compared to that in normal cells, which was related to the inhibition of drug efflux via the ABCB1 transporter down-regulation resulting in delayed retention of intracellular ICG. With artificial intelligence training, the accuracy of image classification into four classes using data sets, such as fluorescence, optical, and fluorescence/optical images was assessed. Fluorescence images obtained the highest accuracy (AUC of 0.8125) than optical and fluorescence/optical images (AUC of 0.75 and 0.6667, respectively). These findings highlight the clinical feasibility of ICG as a detector of precancerous lesions in real-time fluorescence endoscopy with artificial intelligence training and suggest that the mechanism of ICG retention in cancer cells is related to intracellular nitric oxide concentration.
Collapse
Affiliation(s)
- Jinhyeon Kim
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hajung Kim
- Convergence Medicine Research Center, Asan Medical Center, Seoul, Republic of Korea
| | - Yong Sik Yoon
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chan Wook Kim
- Department of Colon and Rectal Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seung-Mo Hong
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sungjee Kim
- Department of Chemistry and School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science & Technology, Pohang, Gyeongbuk, Republic of Korea
| | - Doowon Choi
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science & Technology, Pohang, Gyeongbuk, Republic of Korea
| | - Jihyun Chun
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seung Wook Hong
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung Wook Hwang
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang Hyoung Park
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dong-Hoon Yang
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Byong Duk Ye
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeong-Sik Byeon
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Suk-Kyun Yang
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sun Young Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seung-Jae Myung
- Digestive Diseases Research Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Edis Biotech, Songpa-gu, Seoul, Republic of Korea
| |
Collapse
|
42
|
Wang Y, Nan J, Ma H, Xu J, Guo F, Wang Y, Liang Y, Zhang J, Zhu S. NIR-II Imaging and Sandwiched Plasmonic Biosensor for Ultrasensitive Intraoperative Definition of Tumor-Invaded Lymph Nodes. NANO LETTERS 2023; 23:4039-4048. [PMID: 37071592 PMCID: PMC10176571 DOI: 10.1021/acs.nanolett.3c00829] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Radical lymphadenectomy remains the cornerstone of preventing tumor metastasis through the lymphatic system. Current surgical resection of lymph nodes (LNs) based on fluorescence-guided surgery (FGS) suffers from low sensitivity/selectivity with only qualitative information, hampering accurate intraoperative decision-making. Herein, we develop a modularized theranostic system including NIR-II FGS and a sandwiched plasmonic chip (SPC). Intraoperative NIR-II FGS and detection of tumor-positive lymph nodes were performed on the gastric tumor to determine the feasibility of the modularized theranostic system in defining LN metastasis. Under the NIR-II imaging window, the orthotopic tumor and sentinel lymph nodes (SLNs) were successfully excised without ambient light interference in the operating room. Importantly, the SPC biosensor achieved 100% sensitivity and 100% specificity for tumor markers and realized rapid and high-throughput intraoperative SLN detection. We propose the synergetic design of combining the NIR-II FGS and suitable biosensor will substantially improve the efficiency of cancer diagnosis and therapy follow-up.
Collapse
Affiliation(s)
- Yajun Wang
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Jilin University, Changchun 130021, P.R. China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P.R. China
| | - Jingjie Nan
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Jilin University, Changchun 130021, P.R. China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P.R. China
| | - Huilong Ma
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, P.R. China
| | - Jiajun Xu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Jilin University, Changchun 130021, P.R. China
| | - Feifei Guo
- Cancer Institute, First Hospital of Jilin University, Jilin University, Changchun 130021, P.R. China
| | - Yufeng Wang
- Cancer Institute, First Hospital of Jilin University, Jilin University, Changchun 130021, P.R. China
| | - Yongye Liang
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, P.R. China
| | - Junhu Zhang
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Jilin University, Changchun 130021, P.R. China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P.R. China
| | - Shoujun Zhu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, First Hospital of Jilin University, Jilin University, Changchun 130021, P.R. China
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P.R. China
| |
Collapse
|
43
|
Wang J, Xu Y, Zhang Y, Tian H. Safety and effectiveness of fluorescence laparoscopy in precise hepatectomy: A meta-analysis. Photodiagnosis Photodyn Ther 2023; 42:103599. [PMID: 37156455 DOI: 10.1016/j.pdpdt.2023.103599] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/17/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023]
Abstract
BACKGROUND To perform a systematic review of the safety and effectiveness of fluorescence laparoscopy-guided precise hepatectomy. METHODS We searched the PubMed, Embase, Web of Science, and Cochrane Library databases from inception to December 1, 2022, using the search terms "indocyanine green," "ICG," "infracyanine green," "laparoscopy," "liver resection," and "hepatectomy." After performing a methodological quality assessment of the included studies, the overall results were subjected to meta-analysis using Review Manager 5.3. RESULTS After screening, the meta-analysis included a total of 13 articles. The studies included 1,115 patients who were grouped into the fluorescence laparoscopy (490 patients) and conventional laparoscopy (625 patients) groups. All articles included in the meta-analysis were of high quality. The results of the meta-analysis revealed that compared to the conventional laparoscopy group, the fluorescence laparoscopy group had a higher R0 resection rate (odds ratio=4.03, 95% confidence interval [1.50, 10.83], P=0.006), lower blood transfusion rate (odds ratio=0.46, 95% confidence interval [0.21, 0.97], P=0.04) and lower blood loss (mean difference=-36.58; 95% confidence interval [-59.75, -13.41], P=0.002). However, the length of hospital stay, operative time, and incidence of postoperative complications did not differ significantly between both groups (P>0.05). CONCLUSION Compared to conventional laparoscopy, fluorescence laparoscopy provides better application effects in hepatectomy. The surgical procedure has demonstrated good safety and feasibility, which make it worthy of popularization.
Collapse
Affiliation(s)
- Junqiang Wang
- Shandong University of Traditional Chinese Medicine, Shandong Jinan 250355,China
| | - Ying Xu
- Shandong First Medical University, Shandong Taian 271016, China
| | - Yuhua Zhang
- Shandong University of Traditional Chinese Medicine, Shandong Jinan 250355,China
| | - Hu Tian
- Shandong Provincial Qianfoshan Hospital, Shandong Jinan 250014, China.
| |
Collapse
|
44
|
Roy S, Bag N, Bardhan S, Hasan I, Guo B. Recent Progress in NIR-II Fluorescence Imaging-guided Drug Delivery for Cancer Theranostics. Adv Drug Deliv Rev 2023; 197:114821. [PMID: 37037263 DOI: 10.1016/j.addr.2023.114821] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/20/2023] [Accepted: 04/06/2023] [Indexed: 04/12/2023]
Abstract
Fluorescence imaging in the second near-infrared window (NIR-II) has become a prevalent choice owing to its appealing advantages like deep penetration depth, low autofluorescence, decent spatiotemporal resolution, and a high signal-to-background ratio. This would expedite the innovation of NIR-II imaging-guided drug delivery (IGDD) paradigms for the improvement of the prognosis of patients with tumors. This work systematically reviews the recent progress of such NIR-II IGDD-mediated cancer therapeutics and collectively brings its essence to the readers. Special care has been taken to assess their performances based on their design approach, such as enhancing their drug loading and triggering release, designing intrinsic and extrinsic fluorophores, and/ or overcoming biological barriers. Besides, the state-of-the-art NIR-II IGDD platforms for different therapies like chemo-, photodynamic, photothermal, chemodynamic, immuno-, ion channel, gas-therapies, and multiple functions such as stimulus-responsive imaging and therapy, and monitoring of drug release and therapeutic response, have been updated. In addition, for boosting theranostic outcomes and clinical translation, the innovation directions of NIR-II IGDD platforms are summarized, including renal-clearable, biodegradable, sub-cellular targeting, and/or afterglow, chemiluminescence, X-ray excitable NIR-IGDD, and even cell therapy. This review will propel new directions for safe and efficient NIR-II fluorescence-mediated anticancer drug delivery.
Collapse
Affiliation(s)
- Shubham Roy
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology and School of Science, Harbin Institute of Technology, Shenzhen-518055, China
| | - Neelanjana Bag
- Department of Physics, Jadavpur University, Kolkata-700032, India
| | - Souravi Bardhan
- Department of Physics, Jadavpur University, Kolkata-700032, India
| | - Ikram Hasan
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Bing Guo
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology and School of Science, Harbin Institute of Technology, Shenzhen-518055, China.
| |
Collapse
|
45
|
Price LJ, Tatz J, Sutin J, Spring BQ. Multi-objective optimization of custom compound prism arrays for multiplexed optical imaging. OPTICS EXPRESS 2023; 31:9739-9749. [PMID: 37157537 PMCID: PMC10316679 DOI: 10.1364/oe.475175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/06/2022] [Accepted: 12/11/2022] [Indexed: 05/10/2023]
Abstract
Compound prism arrays are a powerful, yet underutilized, solution for producing high transmission and customized chromatic dispersion profiles over broad bandwidths, the quality of which is unobtainable with commercially available prisms or diffraction gratings. However, the computational complexity associated with designing these prism arrays presents a barrier to the widespread adoption of their use. Here we introduce customizable prism designer software that facilitates high-speed optimization of compound arrays guided by target specifications for chromatic dispersion linearity and detector geometry. Information theory is utilized such that target parameters can be easily modified through user input to efficiently simulate a broad range of possible prism array designs. We demonstrate the capabilities of the designer software to simulate new prism array designs for multiplexed, hyperspectral microscopy that achieve chromatic dispersion linearity and a 70-90% light transmission over a significant portion of the visible wavelength range (500-820 nm). The designer software is applicable to many optical spectroscopy and spectral microscopy applications-with varying requirements for spectral resolution, light ray deviation, and physical size-that are photon-starved and for which the enhanced transmission of refraction versus diffraction warrants custom optical designs.
Collapse
Affiliation(s)
- Liam J. Price
- Department of Physics, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
- Translational Biophotonics Cluster, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Julia Tatz
- Department of Physics, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
- Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Jason Sutin
- Boston Children’s Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Bryan Q. Spring
- Department of Physics, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
- Translational Biophotonics Cluster, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
- Department of Bioengineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| |
Collapse
|
46
|
Hettie KS, Chin FT. NIRDye 812: A molecular platform tailored for multimodal bioimaging applications of targeted fluorescence- and photoacoustic-guided surgery. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2023; 242:112683. [PMID: 36934549 DOI: 10.1016/j.jphotobiol.2023.112683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/16/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023]
Abstract
The primary treatment for malignant tumors remains to be surgical removal of the diseased tissue. The presence or absence of residual diseased tissue at the tumor margin is the strongest predictor of postoperative prognosis and recurrence. Accordingly, reliance on the ability of surgeons to visually distinguish diseased tissue from healthy tissue unambiguously in real time is crucial. Near infrared-I (NIRI) fluorescence-emitting targeting biomolecular constructs such as anticancer antibody-fluorophore conjugates, namely cetuximab-IRDye® 800CW (CTB-IRDye® 800CW), are FDA-approved for clinical trial usage in the fluorescence-guided resection of diseased tissue due to affording improved direct visualization of tumor tissue when compared to the use of either the unaided eye under standard white light illumination (WLI) surgical techniques or non-targeting fluorophores. Unfortunately, though helpful, CTB-IRDye® 800CW affords limited (i) identification of diseased tissue and (ii) tumor margin delineation, because the immunoconjugate generates suboptimal tumor-to-background ratios (TBRs) as a result of its spectral/photophysical profiles poorly aligning with the fixed optical windows of pre-/clinical setups. As such, CTB-IRDye® 800CW is more prone to affording incomplete resection compared to if TBRs were higher due to otherwise. To aid in accurately identifying deep-seated diseased tissue, photoacoustic (PA) tomography has been implemented alongside CTB-IRDye® 800CW to achieve PA signals that could result in higher TBRs. However, in clinical trial practice, using IRDye® 800CW for PA imaging also yields subpar TBRs due to it affording low PA signals. To overcome such limitations, we developed NIRDye 812, a structurally-modified topological equivalent of IRDye® 800CW, to confer it the capability to yield both higher TBRs and superior PA signal than that of the equivalent CTB-conjugate and fluorophore IRDye® 800CW itself, respectively. To do so, we substituted the oxygen atom at its meso-position with a sulfur atom. CTB-NIRDye 812 demonstrated a red-shifted absorption wavelength at 796 nm and a peak NIR-I fluorescence emission wavelength at 820 nm, which better dovetails with the fixed windows of preinstalled fixed emission filters within commercial pre-/clinical NIR-I fluorescence imaging instruments. Overall, CTB-NIRDye 812 provided a ∼ 2-fold increase in TBRs compared to those of CTB-IRDye® 800CW in vivo. Also, NIRDye 812 displayed an ∼60% higher PA signal than that of IRDye® 800CW. Collectively, we achieved our goal of improving upon the spectral/photophysical and PA properties of IRDye® 800CW via introducing a subtle modification to its electronic core such that its CTB immunoconjugate could potentially allow for fast track or breakthrough designation by the FDA due to its near-identical structure displaying considerably improved efficacy.
Collapse
Affiliation(s)
- Kenneth S Hettie
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, 94305, USA; Department of Otolaryngology - Head & Neck Surgery, Stanford University, Stanford, CA 94305, USA.
| | - Frederick T Chin
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
47
|
Chen Y, Streeter SS, Hunt B, Sardar HS, Gunn JR, Tafe LJ, Paydarfar JA, Pogue BW, Paulsen KD, Samkoe KS. Fluorescence molecular optomic signatures improve identification of tumors in head and neck specimens. FRONTIERS IN MEDICAL TECHNOLOGY 2023; 5:1009638. [PMID: 36875185 PMCID: PMC9975724 DOI: 10.3389/fmedt.2023.1009638] [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: 08/02/2022] [Accepted: 01/16/2023] [Indexed: 02/17/2023] Open
Abstract
Background Fluorescence molecular imaging using ABY-029, an epidermal growth factor receptor (EGFR)-targeted, synthetic Affibody peptide labeled with a near-infrared fluorophore, is under investigation for surgical guidance during head and neck squamous cell carcinoma (HNSCC) resection. However, tumor-to-normal tissue contrast is confounded by intrinsic physiological limitations of heterogeneous EGFR expression and non-specific agent uptake. Objective In this preliminary study, radiomic analysis was applied to optical ABY-029 fluorescence image data for HNSCC tissue classification through an approach termed "optomics." Optomics was employed to improve tumor identification by leveraging textural pattern differences in EGFR expression conveyed by fluorescence. The study objective was to compare the performance of conventional fluorescence intensity thresholding and optomics for binary classification of malignant vs. non-malignant HNSCC tissues. Materials and Methods Fluorescence image data collected through a Phase 0 clinical trial of ABY-029 involved a total of 20,073 sub-image patches (size of 1.8 × 1.8 mm2) extracted from 24 bread-loafed slices of HNSCC surgical resections originating from 12 patients who were stratified into three dose groups (30, 90, and 171 nanomoles). Each dose group was randomly partitioned on the specimen-level 75%/25% into training/testing sets, then all training and testing sets were aggregated. A total of 1,472 standardized radiomic features were extracted from each patch and evaluated by minimum redundancy maximum relevance feature selection, and 25 top-ranked features were used to train a support vector machine (SVM) classifier. Predictive performance of the SVM classifier was compared to fluorescence intensity thresholding for classifying testing set image patches with histologically confirmed malignancy status. Results Optomics provided consistent improvement in prediction accuracy and false positive rate (FPR) and similar false negative rate (FNR) on all testing set slices, irrespective of dose, compared to fluorescence intensity thresholding (mean accuracies of 89% vs. 81%, P = 0.0072; mean FPRs of 12% vs. 21%, P = 0.0035; and mean FNRs of 13% vs. 17%, P = 0.35). Conclusions Optomics outperformed conventional fluorescence intensity thresholding for tumor identification using sub-image patches as the unit of analysis. Optomics mitigate diagnostic uncertainties introduced through physiological variability, imaging agent dose, and inter-specimen biases of fluorescence molecular imaging by probing textural image information. This preliminary study provides a proof-of-concept that applying radiomics to fluorescence molecular imaging data offers a promising image analysis technique for cancer detection in fluorescence-guided surgery.
Collapse
Affiliation(s)
- Yao Chen
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| | - Samuel S. Streeter
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
- Department of Orthopaedics, Dartmouth Health, Lebanon, NH, United States
- Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
| | - Brady Hunt
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| | - Hira S. Sardar
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| | - Jason R. Gunn
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| | - Laura J. Tafe
- Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
- Department of Pathology, Dartmouth Health, Lebanon, NH, United States
| | - Joseph A. Paydarfar
- Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
- Department of Surgery, Dartmouth Health, Lebanon, NH, United States
- Department of Otolaryngology, Dartmouth Health, Lebanon, NH, United States
| | - Brian W. Pogue
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
- Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, United States
| | - Keith D. Paulsen
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
- Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
| | - Kimberley S. Samkoe
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
- Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
- Department of Surgery, Dartmouth Health, Lebanon, NH, United States
| |
Collapse
|
48
|
Wang C, Xu X, Hodge S, Chen EY, Hoopes PJ, Tichauer KM, Samkoe KS. Identification of a Suitable Untargeted Agent for the Clinical Translation of ABY-029 Paired-Agent Imaging in Fluorescence-Guided Surgery. Mol Imaging Biol 2023; 25:97-109. [PMID: 34642897 PMCID: PMC9413473 DOI: 10.1007/s11307-021-01642-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/05/2021] [Accepted: 08/12/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE Non-specific uptake and retention of molecular targeted agents and heterogeneous tissue optical properties diminish the ability to differentiate between tumor and normal tissues using molecular targeted fluorescent agents. Paired-agent imaging (PAI) can increase the diagnostic ability to detect tumor tissue by mitigating these non-specific effects and providing true molecular contrast by co-administration of an untargeted control imaging agent with a targeted agent. This study evaluates the suitability of available clinically translatable untargeted agents for the translation of PAI in fluorescence-guided surgery using an affibody-based targeted imaging agent (ABY-029). EXPERIMENTAL DESIGN: Three untargeted agents that fluoresce near 700 nm and exhibit good clinical safety profiles (methylene blue, IRDye 700DX, and IRDye 680LT) were tested in combination with the clinically tested IRDye 800CW-labeled anti-epidermal growth factor receptor (EGFR) affibody molecule, ABY-029 (eIND 122,681). Properties of the untargeted agent important for human use and integrity of PAI were tested: (1) plasma protein binding; (2) fluorescence signal linearity in in vitro whole blood dilution; (3) in vivo pharmacokinetic matching to targeted agent in negative control tissue; and (4) in vivo diagnostic accuracy of PAI vs single agent imaging (SAI) of ABY-029 alone in orthotopic oral head and neck squamous cell carcinomas. RESULTS IRDye 680LT outperformed IRDye 700DX and methylene blue with the highest signal linearity (R2 = 0.9998 ± 0.0002, 0.9995 ± 0.0004, 0.91 ± 0.02, respectively), the highest fluorescence yield in whole blood at 1 μM (104.42 ± 0.05, 103.68 ± 0.09, 101.9 ± 0.2, respectively), and the most closely matched ABY-029 pharmacokinetics in EGFR-negative tissues (binding potential error percentage = 0.31% ± 0.37%, 10.25% ± 1.30%, and 8.10% ± 5.37%, respectively). The diagnostic ability of PAI with ABY-029 and IRDye 680LT outperformed conventional SAI with an area-under-the-receiver-operating-characteristic curve (AUC) value of 0.964 vs. 0.854, and 0.978 vs. 0.925 in the Odyssey scanning system and Pearl wide field imaging system, respectively. CONCLUSION PAI is a highly promising methodology for increasing detection of tumors in fluorescence-guided surgery. Although not yet clinically approved, IRDye 680LT demonstrates promise as an untargeted agent when paired with ABY-029. The clinical translation of PAI to maximize tumor excision, while minimizing normal tissue removal, could improve both patient survival and life quality.
Collapse
Affiliation(s)
- Cheng Wang
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Xiaochun Xu
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Sassan Hodge
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Eunice Y Chen
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - P Jack Hoopes
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA.,Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Kenneth M Tichauer
- Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA
| | - Kimberley S Samkoe
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA. .,Geisel School of Medicine, Dartmouth College, Hanover, NH, USA.
| |
Collapse
|
49
|
Van Keulen S, Hom M, White H, Rosenthal EL, Baik FM. The Evolution of Fluorescence-Guided Surgery. Mol Imaging Biol 2023; 25:36-45. [PMID: 36123445 PMCID: PMC9971137 DOI: 10.1007/s11307-022-01772-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 10/14/2022]
Abstract
There has been continual development of fluorescent agents, imaging systems, and their applications over the past several decades. With the recent FDA approvals of 5-aminolevulinic acid, hexaminolevulinate, and pafolacianine, much of the potential that fluorescence offers for image-guided oncologic surgery is now being actualized. In this article, we review the evolution of fluorescence-guided surgery, highlight the milestones which have contributed to successful clinical translation, and examine the future of targeted fluorescence imaging.
Collapse
Affiliation(s)
- Stan Van Keulen
- Department of Oral and Maxillofacial Surgery and Oral Pathology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Marisa Hom
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Haley White
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eben L Rosenthal
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Fred M Baik
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
50
|
Li T, Cao K, Yang X, Liu Y, Wang X, Wu F, Chen G, Wang Q. An oral ratiometric NIR-II fluorescent probe for reliable monitoring of gastrointestinal diseases in vivo. Biomaterials 2023; 293:121956. [PMID: 36543049 DOI: 10.1016/j.biomaterials.2022.121956] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/09/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
Early monitoring of gastrointestinal diseases via orally delivered NIR-II ratiometric fluorescent probes represents a promising noninvasive diagnostic modality, but is challenging due to the limitation of harsh digestive environment. Here, we report a single-component NIR-II ratiometric molecular nanoprobe (LC-1250 NP) to monitor gastrointestinal disease with high specificity to its biomarker H2O2 via oral administration. LC-1250 NP displays stable fluorescence in the channel of 1250 long-pass (F1250LP) before and after the gastrointestinal disease detection as the reference, while it presents significantly enhanced fluorescence signal in the response channel of 1150 nm short-pass (F1150SP) in diseased gastrointestinal environment due to the intramolecular cyclization of LC-1250 molecules activated by H2O2. The fluorescence ratio (F1150SP/F1250LP) increases linearly with the concentration of H2O2 with a low detection limit of 20 nM. Therefore, when delivered orally, LC-1250 NP can accurately map the diseased areas and surmount the false-positive interference from biological heterogeneity by NIR-II ratiometric fluorescence imaging, providing sensitive and reliable evaluation for the progress of gastroenteritis.
Collapse
Affiliation(s)
- Tuanwei Li
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Kaili Cao
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Xiaohu Yang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Yongyang Liu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Xingyu Wang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Feng Wu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Guangcun Chen
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China.
| | - Qiangbin Wang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Key Laboratory of Functional Molecular Imaging Technology, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China; School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China; College of Materials Sciences and Opto-Electronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|