1
|
Zhang T, Yang B, Jiang T, Kong X, Huo X, Ma Y, Yang K, Liu M, Liu Y, Yao Z, Yu H, Liu H, Zhang K, Liu Y. A Hypoxia-Activated BODIPY-Azo Anticancer Prodrug for Bimodal Chemo-Photodynamic Therapy. J Med Chem 2025. [PMID: 39826133 DOI: 10.1021/acs.jmedchem.4c02231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
For cancer treatment, collaborative strategies have been the mainstream for overcoming the restrictions resulting from monotherapy. Combining chemotherapy with photodynamic therapy (PDT) has been shown to increase the antitumor effect and reduce side impacts. This study reports a hypoxia-activated prodrug BOD-Azo-single with a PDT agent and aniline mustard connected by the azo bond. With light illumination, BOD-Azo-single exhibited some PDT. Under hypoxic conditions, the azo bond cleaved and released BOD-3-single of higher phototoxicity and aniline mustard of chemotoxicity. In vivo therapeutic experiments showed that BOD-Azo-single with light significantly reduced A375 tumor proliferation with 92% TGI value. Overall, in this study, PDT was employed to address the adverse systemic toxicity of chemotherapy and the released chemotoxicity made up for the inefficiency of PDT in the hypoxic tumor microenvironment, introducing a new strategy for developing combined therapeutic agents to be advantageous to each other. Under a hypoxic tumor environment, BOD-3-single and aniline mustard exerted a strong synergistic effect (CI = 0.25), indicating that BOD-Azo-single is a real bimodal chemo-photodynamic therapeutic agent.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Medicinal Chemistry, Hebei Medical University, Shijiazhuang 050017, China
| | - Baoyin Yang
- Department of Pharmacy, Baoding Hospital of Beijing Children's Hospital, Capital Medical University, Baoding 071000, China
| | - Tao Jiang
- The Thoracic Surgery Department of the First Hospital of Hebei Medical University, Shijiazhuang 050017, China
| | - Xiangyu Kong
- Department of Medicinal Chemistry, Hebei Medical University, Shijiazhuang 050017, China
| | - Xinyao Huo
- Department of Medicinal Chemistry, Hebei Medical University, Shijiazhuang 050017, China
| | - Yan Ma
- Department of Medicinal Chemistry, Hebei Medical University, Shijiazhuang 050017, China
| | - Kehao Yang
- Department of Medicinal Chemistry, Hebei Medical University, Shijiazhuang 050017, China
| | - Mengchun Liu
- Renhe Yikang Group Co., Ltd, Shijiazhuang 050017, China
| | - Yumiao Liu
- Renhe Yikang Group Co., Ltd, Shijiazhuang 050017, China
| | - Zikuo Yao
- Department of Medicinal Chemistry, Hebei Medical University, Shijiazhuang 050017, China
| | - Hao Yu
- Department of Medicinal Chemistry, Hebei Medical University, Shijiazhuang 050017, China
| | - Huining Liu
- The Thoracic Surgery Department of the First Hospital of Hebei Medical University, Shijiazhuang 050017, China
| | - Kai Zhang
- Department of Medicinal Chemistry, Hebei Medical University, Shijiazhuang 050017, China
| | - Yifan Liu
- Department of Medicinal Chemistry, Hebei Medical University, Shijiazhuang 050017, China
| |
Collapse
|
2
|
Srinivasan D, Subbarayan R, Krishnan M, Balakrishna R, Adtani P, Shrestha R, Chauhan A, Babu S, Radhakrishnan A. Radiation therapy-induced normal tissue damage: involvement of EMT pathways and role of FLASH-RT in reducing toxicities. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2025:10.1007/s00411-024-01102-2. [PMID: 39760753 DOI: 10.1007/s00411-024-01102-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 12/20/2024] [Indexed: 01/07/2025]
Abstract
Radiation therapy (RT) is fundamental to the fight against cancer because of its exceptional ability to target and destroy cancer cells. However, conventional radiation therapy can significantly affect the adjacent normal tissues, leading to fibrosis, inflammation, and decreased organ function. This tissue damage not only reduces the quality of life but also prevents the total elimination of cancer. The transformation of epithelial cells into mesenchymal-like cells, termed epithelial-mesenchymal transition (EMT), is essential for processes such as fibrosis, embryogenesis, and wound healing. Conventional radiation therapy increases the asymmetric activation of fibrotic and inflammatory pathways, and the resulting chronic fibrotic changes and organ dysfunction are linked to radiation-induced epithelial-mesenchymal transition. Recent advances in radiation therapy, namely flash radiation therapy (FLASH-RT), have the potential to widen the therapeutic index. Radiation delivered by FLASH-RT at very high dose rates (exceeding 40 Gy/s) can protect normal tissue from radiation-induced damage, a phenomenon referred to as the "FLASH effect". Preclinical studies have demonstrated that FLASH-RT successfully inhibits processes associated with fibrosis and epithelial-mesenchymal transition, mitigates damage to normal tissue, and enhances regeneration. Three distinct types of EMT have been identified: type-1, associated with embryogenesis; Type-2, associated with injury potential; and type-3, related with cancer spread. The regulation of EMT via pathways, including TGF-β/SMAD, WNT/β-catenin, and NF-κB, is essential for radiation-induced tissue remodelling. This study examined radiation-induced EMT, TGF-β activity, multiple signalling pathways in fibrosis, and the potential of FLASH-RT to reduce tissue damage. FLASH-RT is a novel approach to treat chronic tissue injury and fibrosis post-irradiation by maintaining epithelial properties and regulating mesenchymal markers including vimentin and N-cadherin. Understanding these pathways will facilitate the development of future therapies that can alleviate fibrosis, improve the efficacy of cancer therapy, and improve the quality of life of patients.
Collapse
Affiliation(s)
- Dhasarathdev Srinivasan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103, Tamil Nadu, India
| | - Rajasekaran Subbarayan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103, Tamil Nadu, India.
- Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103, Tamil Nadu, India.
| | - Madhan Krishnan
- Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103, Tamil Nadu, India
| | - Ranjith Balakrishna
- Centre for Advanced Biotherapeutics and Regenerative Medicine, Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103, Tamil Nadu, India
| | - Pooja Adtani
- Department of Basic Medical and Dental Sciences, Gulf Medical University, Ajman, United Arab Emirates
| | - Rupendra Shrestha
- Department of Natural and Applied Sciences, Nexus Institute of Research and Innovation (NIRI), Lalitpur, Nepal.
| | - Ankush Chauhan
- Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103, Tamil Nadu, India
| | - Shyamaladevi Babu
- Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103, Tamil Nadu, India
| | - Arunkumar Radhakrishnan
- Department of Pharmacology, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| |
Collapse
|
3
|
Qi L, Li Z, Liu J, Chen X. Omics-Enhanced Nanomedicine for Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409102. [PMID: 39473316 DOI: 10.1002/adma.202409102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/10/2024] [Indexed: 12/13/2024]
Abstract
Cancer nanomedicine has emerged as a promising approach to overcome the limitations of conventional cancer therapies, offering enhanced efficacy and safety in cancer management. However, the inherent heterogeneity of tumors presents increasing challenges for the application of cancer nanomedicine in both diagnosis and treatment. This heterogeneity necessitates the integration of advanced and high-throughput analytical techniques to tailor nanomedicine strategies to individual tumor profiles. Omics technologies, encompassing genomics, epigenomics, transcriptomics, proteomics, metabolomics, and more, provide unparalleled insights into the molecular and cellular mechanisms underlying cancer. By dissecting tumor heterogeneity across multiple levels, these technologies offer robust support for the development of personalized and precise cancer nanomedicine strategies. In this review, the principles, techniques, and applications of key omics technologies are summarized. Especially, the synergistic integration of omics and nanomedicine in cancer therapy is explored, focusing on enhanced diagnostic accuracy, optimized therapeutic strategies and the assessment of nanomedicine-mediated biological responses. Moreover, this review addresses current challenges and outlines future directions in the field of omics-enhanced nanomedicine. By offering valuable insights and guidance, this review aims to advance the integration of omics with nanomedicine, ultimately driving improved diagnostic and therapeutic strategies for cancer.
Collapse
Affiliation(s)
- Lin Qi
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, Hunan, 410011, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Zhihong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, Hunan, 410011, China
| | - Jianping Liu
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Xiaoyuan Chen
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, Hunan, 410011, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore, 138667, Singapore
| |
Collapse
|
4
|
Zang X, Lei K, Wang J, Gong R, Gao C, Jing Z, Song J, Ren H. Targeting aberrant amino acid metabolism for pancreatic cancer therapy: Opportunities for nanoparticles. CHEMICAL ENGINEERING JOURNAL 2024; 498:155071. [DOI: 10.1016/j.cej.2024.155071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
5
|
Ahmad F, Muhmood T. Clinical translation of nanomedicine with integrated digital medicine and machine learning interventions. Colloids Surf B Biointerfaces 2024; 241:114041. [PMID: 38897022 DOI: 10.1016/j.colsurfb.2024.114041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 06/21/2024]
Abstract
Nanomaterials based therapeutics transform the ways of disease prevention, diagnosis and treatment with increasing sophistications in nanotechnology at a breakneck pace, but very few could reach to the clinic due to inconsistencies in preclinical studies followed by regulatory hinderances. To tackle this, integrating the nanomedicine discovery with digital medicine provide technologies as tools of specific biological activity measurement. Hence, overcome the redundancies in nanomedicine discovery by the on-site data acquisition and analytics through integrating intelligent sensors and artificial intelligence (AI) or machine learning (ML). Integrated AI/ML wearable sensors directly gather clinically relevant biochemical information from the subject's body and process data for physicians to make right clinical decision(s) in a time and cost-effective way. This review summarizes insights and recommend the infusion of actionable big data computation enabled sensors in burgeoning field of nanomedicine at academia, research institutes, and pharmaceutical industries, with a potential of clinical translation. Furthermore, many blind spots are present in modern clinically relevant computation, one of which could prevent ML-guided low-cost new nanomedicine development from being successfully translated into the clinic was also discussed.
Collapse
Affiliation(s)
- Farooq Ahmad
- State Key Laboratory of Chemistry and Utilization of Carbon Based Energy Resources, College of Chemistry, Xinjiang University, Urumqi 830017, China.
| | - Tahir Muhmood
- International Iberian Nanotechnology Laboratory (INL), Avenida Mestre José Veiga, Braga 4715-330, Portugal.
| |
Collapse
|
6
|
Liang G, Cao W, Tang D, Zhang H, Yu Y, Ding J, Karges J, Xiao H. Nanomedomics. ACS NANO 2024; 18:10979-11024. [PMID: 38635910 DOI: 10.1021/acsnano.3c11154] [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: 04/20/2024]
Abstract
Nanomaterials have attractive physicochemical properties. A variety of nanomaterials such as inorganic, lipid, polymers, and protein nanoparticles have been widely developed for nanomedicine via chemical conjugation or physical encapsulation of bioactive molecules. Superior to traditional drugs, nanomedicines offer high biocompatibility, good water solubility, long blood circulation times, and tumor-targeting properties. Capitalizing on this, several nanoformulations have already been clinically approved and many others are currently being studied in clinical trials. Despite their undoubtful success, the molecular mechanism of action of the vast majority of nanomedicines remains poorly understood. To tackle this limitation, herein, this review critically discusses the strategy of applying multiomics analysis to study the mechanism of action of nanomedicines, named nanomedomics, including advantages, applications, and future directions. A comprehensive understanding of the molecular mechanism could provide valuable insight and therefore foster the development and clinical translation of nanomedicines.
Collapse
Affiliation(s)
- Ganghao Liang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Wanqing Cao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei 230026, P. R. China
| | - Dongsheng Tang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Hanchen Zhang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yingjie Yu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei 230026, P. R. China
| | - Johannes Karges
- Faculty of Chemistry and Biochemistry, Ruhr-University Bochum, Universitätsstrasse 150, 44780 Bochum, Germany
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
7
|
Tsang CY, Zhang Y. Nanomaterials for light-mediated therapeutics in deep tissue. Chem Soc Rev 2024; 53:2898-2931. [PMID: 38265834 DOI: 10.1039/d3cs00862b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
Light-mediated therapeutics, including photodynamic therapy, photothermal therapy and light-triggered drug delivery, have been widely studied due to their high specificity and effective therapy. However, conventional light-mediated therapies usually depend on the activation of light-sensitive molecules with UV or visible light, which have poor penetration in biological tissues. Over the past decade, efforts have been made to engineer nanosystems that can generate luminescence through excitation with near-infrared (NIR) light, ultrasound or X-ray. Certain nanosystems can even carry out light-mediated therapy through chemiluminescence, eliminating the need for external activation. Compared to UV or visible light, these 4 excitation modes penetrate more deeply into biological tissues, triggering light-mediated therapy in deeper tissues. In this review, we systematically report the design and mechanisms of different luminescent nanosystems excited by the 4 excitation sources, methods to enhance the generated luminescence, and recent applications of such nanosystems in deep tissue light-mediated therapeutics.
Collapse
Affiliation(s)
- Chung Yin Tsang
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117583, Singapore.
| | - Yong Zhang
- Department of Biomedical Engineering, The City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong.
| |
Collapse
|
8
|
Ma G, Zhang X, Zhao K, Zhang S, Ren K, Mu M, Wang C, Wang X, Liu H, Dong J, Sun X. Polydopamine Nanostructure-Enhanced Water Interaction with pH-Responsive Manganese Sulfide Nanoclusters for Tumor Magnetic Resonance Contrast Enhancement and Synergistic Ferroptosis-Photothermal Therapy. ACS NANO 2024; 18:3369-3381. [PMID: 38251846 DOI: 10.1021/acsnano.3c10249] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Rational structure design benefits the development of efficient nanoplatforms for tumor theranostic application. In this work, a multifunctional polydopamine (PDA)-coated manganese sulfide (MnS) nanocluster was prepared. The polyhydroxy structure of PDA enhanced the water interaction with pH-responsive MnS nanoclusters via hydrogen bonds. At pH 5.5 conditions, the spin-lattice relaxation rate of MnS nanoclusters dramatically increased from 5.76 to 19.33 mM-1·s-1 after the PDA coating, which can be beneficial for efficient tumor magnetic resonance imaging. In addition, PDA endowed MnS nanoclusters with excellent biocompatibility and good photothermal conversion efficiency, which can be used for efficient tumor photothermal therapy (PTT). Furthermore, MnS nanoclusters possess the ability to release H2S in the acidic tumor microenvironment, effectively inhibiting mitochondrial respiration and adenosine triphosphate production. As a result, the expression of heat shock protein was obviously reduced, which can reduce the resistance of tumor cells to photothermal stimulation and enhance the efficacy of PTT. The released Mn2+ also displayed efficient peroxidase and glutathione oxidase-like activity, effectively inducing tumor cell ferroptosis and apoptosis at the same time. Therefore, this nanoplatform could be a potential nanotheranostic for magnetic resonance contrast enhancement and synergistic ferroptosis-PTT of tumors.
Collapse
Affiliation(s)
- Guiqi Ma
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Xinyu Zhang
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Kunlong Zhao
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Shuxuan Zhang
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Ke Ren
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Mengyao Mu
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Chenyu Wang
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Ximing Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Hui Liu
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Jian Dong
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250000, China
| | - Xiao Sun
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250000, China
- Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, China
| |
Collapse
|
9
|
Yao B, Liu X, Zhang W, Lu H. X-ray excited luminescent nanoparticles for deep photodynamic therapy. RSC Adv 2023; 13:30133-30150. [PMID: 37849702 PMCID: PMC10577683 DOI: 10.1039/d3ra04984a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 09/22/2023] [Indexed: 10/19/2023] Open
Abstract
Photodynamic therapy (PDT), as a non-invasive treatment, has received wide attention because of its high selectivity and low side effects. However, traditional PDT is influenced by the excitation light source and the light penetration depth is limited, which can only be used for superficial epidermal tumor treatment, and it is still a great challenge for deep tumor treatment. In recent years, X-ray excitation photodynamic therapy (X-PDT) using penetrating X-rays as an external excitation source and X-ray excited luminescent nanoparticles (XLNP) as an energy transfer medium to indirectly excite photosensitizer (PS) has solved the problem of insufficient penetration depth in tissues and become a research hotspot in the field of deep tumor treatment. In this review, the recent research progress of nanoparticles for efficient X-PDT, listing different types of XLNP and luminescence enhancement strategies. The loading method of PS is highlighted to achieve efficient energy transfer by regulating the intermolecular distance between both XLNP/PS. In addition, the water-soluble modification of XLNP surface is discussed and different hydrophilic modification methods are proposed to provide reference ideas for improving the dispersibility and biocompatibility of XLNP in aqueous solution. Finally, the therapeutic effects about X-PDT are discussed, and the current challenges and future perspectives for its clinical applications are presented.
Collapse
Affiliation(s)
- Bang Yao
- School of Materials Science and Engineering, Shaanxi University of Science and Technology Xian 710021 China
- School of Biomedical Engineering, Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, The Fourth Military Medical University 169th Changle West Road Xi'an Shaanxi 710032 China
| | - Xiaoxu Liu
- School of Materials Science and Engineering, Shaanxi University of Science and Technology Xian 710021 China
| | - Wenli Zhang
- School of Biomedical Engineering, Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, The Fourth Military Medical University 169th Changle West Road Xi'an Shaanxi 710032 China
| | - Hongbing Lu
- School of Biomedical Engineering, Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, The Fourth Military Medical University 169th Changle West Road Xi'an Shaanxi 710032 China
| |
Collapse
|
10
|
Mandl GA, Vettier F, Tessitore G, Maurizio SL, Bietar K, Stochaj U, Capobianco JA. Combining Pr 3+-Doped Nanoradiosensitizers and Endogenous Protoporphyrin IX for X-ray-Mediated Photodynamic Therapy of Glioblastoma Cells. ACS APPLIED BIO MATERIALS 2023. [PMID: 37267436 DOI: 10.1021/acsabm.3c00201] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Glioblastoma multiforme is an aggressive type of brain cancer with high recurrence rates due to the presence of radioresistant cells remaining after tumor resection. Here, we report the development of an X-ray-mediated photodynamic therapy (X-PDT) system using NaLuF4:25% Pr3+ radioluminescent nanoparticles in conjunction with protoporphyrin IX (PPIX), an endogenous photosensitizer that accumulates selectively in cancer cells. Conveniently, 5-aminolevulinic acid (5-ALA), the prodrug that is administered for PDT, is the only drug approved for fluorescence-guided resection of glioblastoma, enabling dual detection and treatment of malignant cells. NaLuF4:Pr3+ nanoparticles were synthesized and spectroscopically evaluated at a range of Pr3+ concentrations. This generated radioluminescent nanoparticles with strong emissions from the 1S0 excited state of Pr3+, which overlaps with the Soret band of PPIX to perform photodynamic therapy. The spectral overlap between the nanoparticles and PPIX improved treatment outcomes for U251 cells, which were used as a model for the thin tumor margin. In addition to sensitizing PPIX to induce X-PDT, our nanoparticles exhibit strong radiosensitizing properties through a radiation dose-enhancement effect. We evaluate the effects of the nanoparticles alone and in combination with PPIX on viability, death, stress, senescence, and proliferation. Collectively, our results demonstrate this as a strong proof of concept for nanomedicine.
Collapse
Affiliation(s)
- Gabrielle A Mandl
- Department of Chemistry and Biochemistry & Centre for Nanoscience Research, Concordia University, 7141 Sherbrooke St. W., Montreal, Quebec H4B 1R6, Canada
| | - Freesia Vettier
- Department of Chemistry and Biochemistry & Centre for Nanoscience Research, Concordia University, 7141 Sherbrooke St. W., Montreal, Quebec H4B 1R6, Canada
| | - Gabriella Tessitore
- Department of Chemistry and Biochemistry & Centre for Nanoscience Research, Concordia University, 7141 Sherbrooke St. W., Montreal, Quebec H4B 1R6, Canada
| | - Steven L Maurizio
- Department of Chemistry and Biochemistry & Centre for Nanoscience Research, Concordia University, 7141 Sherbrooke St. W., Montreal, Quebec H4B 1R6, Canada
| | - Kais Bietar
- Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Ursula Stochaj
- Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - John A Capobianco
- Department of Chemistry and Biochemistry & Centre for Nanoscience Research, Concordia University, 7141 Sherbrooke St. W., Montreal, Quebec H4B 1R6, Canada
| |
Collapse
|
11
|
Secchi V, Cova F, Villa I, Babin V, Nikl M, Campione M, Monguzzi A. Energy Partitioning in Multicomponent Nanoscintillators for Enhanced Localized Radiotherapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:24693-24700. [PMID: 37172016 DOI: 10.1021/acsami.3c00853] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Multicomponent nanomaterials consisting of dense scintillating particles functionalized by or embedding optically active conjugated photosensitizers (PSs) for cytotoxic reactive oxygen species (ROS) have been proposed in the last decade as coadjuvant agents for radiotherapy of cancer. They have been designed to make scintillation-activated sensitizers for ROS production in an aqueous environment under exposure to ionizing radiations. However, a detailed understanding of the global energy partitioning process occurring during the scintillation is still missing, in particular regarding the role of the non-radiative energy transfer between the nanoscintillator and the conjugated moieties which is usually considered crucial for the activation of PSs and therefore pivotal to enhance the therapeutic effect. We investigate this mechanism in a series of PS-functionalized scintillating nanotubes where the non-radiative energy transfer yield has been tuned by control of the intermolecular distance between the nanotube and the conjugated system. The obtained results indicate that non-radiative energy transfer has a negligible effect on the ROS sensitization efficiency, thus opening the way to the development of different architectures for breakthrough radiotherapy coadjutants to be tested in clinics.
Collapse
Affiliation(s)
- Valeria Secchi
- Dipartimento di Scienza Dei Materiali, Università Degli Studi Milano-Bicocca, 20125 Milano, Italy
- NANOMIB, Center for Biomedical Nanomedicine, University of Milano-Bicocca, P.zza Ateneo Nuovo 1, 20126 Milan, Italy
| | - Francesca Cova
- Dipartimento di Scienza Dei Materiali, Università Degli Studi Milano-Bicocca, 20125 Milano, Italy
| | - Irene Villa
- Dipartimento di Scienza Dei Materiali, Università Degli Studi Milano-Bicocca, 20125 Milano, Italy
- FZU─Institute of Physics of the Czech Academy of Sciences, Cukrovarnická 10/112, 16 200 Prague, Czech Republic
| | - Vladimir Babin
- FZU─Institute of Physics of the Czech Academy of Sciences, Cukrovarnická 10/112, 16 200 Prague, Czech Republic
| | - Martin Nikl
- FZU─Institute of Physics of the Czech Academy of Sciences, Cukrovarnická 10/112, 16 200 Prague, Czech Republic
| | - Marcello Campione
- NANOMIB, Center for Biomedical Nanomedicine, University of Milano-Bicocca, P.zza Ateneo Nuovo 1, 20126 Milan, Italy
- Department of Earth and Environmental Sciences, Università Degli Studi Milano-Bicocca, Piazza Della Scienza 4, 20126 Milano, Italy
| | - Angelo Monguzzi
- Dipartimento di Scienza Dei Materiali, Università Degli Studi Milano-Bicocca, 20125 Milano, Italy
- NANOMIB, Center for Biomedical Nanomedicine, University of Milano-Bicocca, P.zza Ateneo Nuovo 1, 20126 Milan, Italy
| |
Collapse
|
12
|
Gu X, Shu T, Deng W, Shen C, Wu Y. An X-ray activatable gold nanorod encapsulated liposome delivery system for mitochondria-targeted photodynamic therapy (PDT). J Mater Chem B 2023; 11:4539-4547. [PMID: 37161717 DOI: 10.1039/d3tb00608e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
In this work, we developed a mitochondria-targeted nanomaterial for neoadjuvant X-ray-triggered photodynamic therapy of rectal cancer. Herein, we designed a biodegradable liposome incorporating a photosensitizer, verteporfin, to generate X-ray-induced reactive oxygen species, gold nanorods as radiation enhancers, and triphenylphosphonium as the mitochondrial targeting moiety. The average size of the nanocarrier was about 150 nm. Due to the synergetic effect between X-ray and a combination of verteporfin and gold nanorods, as well as precise site-targeted TPP-modified liposomal nanocarriers, our nanoconjugates generated sufficient cytotoxic singlet oxygen within the mitochondria under X-ray irradiation, triggering the loss of membrane potential and mitochondria-related apoptosis of cancer cells.
Collapse
Affiliation(s)
- Xuefan Gu
- College of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, Shaanxi, 710065, P. R. China
- ARC Centre of Excellence for Nanoscale Biophotonics, Graduate School of Biomedical Engineering, University of New South, Wales Kensington, 2052 NSW, Australia
- Faculty of Science and Engineering, Macquarie University, Sydney, 2109 NSW, Australia
| | - Tiantian Shu
- College of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, Shaanxi, 710065, P. R. China
| | - Wei Deng
- School of Biomedical Engineering, University of Technology Sydney, Sydney, Australia
| | - Chao Shen
- Faculty of Science and Engineering, Macquarie University, Sydney, 2109 NSW, Australia
| | - Youshen Wu
- Department of Chemistry, School of Science, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China.
| |
Collapse
|
13
|
Jiang X, Gao X, Li L, Zhou P, Wang S, Liu T, Zhou J, Zhang H, Huang K, Li Y, Wang M, Jin Z, Xie E, Liu W, Han G. Enhancement of Light and X-ray Charging in Persistent Luminescence Nanoparticle Scintillators Zn 2SiO 4:Mn 2+, Yb 3+, Li . ACS APPLIED MATERIALS & INTERFACES 2023; 15:21228-21238. [PMID: 37078901 DOI: 10.1021/acsami.3c00664] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Persistent luminescence nanoparticle scintillators (PLNS) have been attempted for X-ray-induced photodynamic therapy (X-PDT) because persistent luminescence after ceasing radiation can make PLNS use less cumulative irradiation time and dose to generate the same amount of reactive oxygen species (ROS) compared with conventional scintillators to combat cancer cells. However, excessive surface defects in PLNS reduce the luminescence efficiency and quench the persistent luminescence, which is fatal to the efficacy of X-PDT. Herein, the PLNS of SiO2@Zn2SiO4:Mn2+, Yb3+, Li+ was designed by the energy trap engineering and synthesized by a simple template method, which has excellent X-ray and UV-excited persistent luminescence and continuously tunable emission spectra from 520 to 550 nm. Its luminescence intensity and afterglow time are more than 7 times that of the reported Zn2SiO4:Mn2+ used for X-PDT. By loading a Rose Bengal (RB) photosensitizer, an effective persistent energy transfer from the PLNS to photosensitizer is observed even after the removal of X-ray irradiation. The X-ray dose of nanoplatform SiO2@Zn2SiO4:Mn2+, Yb3+, Li+@RB in X-PDT of HeLa cancer cells was reduced to 0.18 Gy compared to the X-ray dose of 1.0 Gy for Zn2SiO4:Mn for X-PDT. This indicates that the Zn2SiO4:Mn2+, Yb3+, Li+ PLNS have great potential for X-PDT applications.
Collapse
Affiliation(s)
- Xiaohui Jiang
- School of Physical Science and Technology, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Xiuping Gao
- School of Physical Science and Technology, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Lingyi Li
- The High School Attached to Northwest Normal University, Lanzhou, Gansu 730000, P. R. China
| | - Ping Zhou
- School of Stomatology, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Shasha Wang
- School of Physical Science and Technology, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Tao Liu
- School of Physical Science and Technology, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Jinyuan Zhou
- School of Physical Science and Technology, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Haodong Zhang
- School of Physical Science and Technology, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Kai Huang
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| | - Yang Li
- School of Basic Medicine, Guangzhou Medical University, Guangzhou, Guangdong 511436, P. R. China
| | - Min Wang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Zhiwen Jin
- School of Physical Science and Technology, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Erqing Xie
- School of Physical Science and Technology, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Weisheng Liu
- Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province and State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, P. R. China
| | - Gang Han
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, United States
| |
Collapse
|
14
|
Zhang G, Guo M, Ma H, Wang J, Zhang XD. Catalytic nanotechnology of X-ray photodynamics for cancer treatments. Biomater Sci 2023; 11:1153-1181. [PMID: 36602259 DOI: 10.1039/d2bm01698b] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Photodynamic therapy (PDT) has been applied in cancer treatment because of its high selectivity, low toxicity, and non-invasiveness. However, the limited penetration depth of the light still hampers from reaching deep-seated tumors. Considering the penetrating ability of high-energy radiotherapy, X-ray-induced photodynamic therapy (X-PDT) has evolved as an alternative to overcome tissue blocks. As the basic principle of X-PDT, X-rays stimulate the nanoparticles to emit scintillating or persistent luminescence and further activate the photosensitizers to generate reactive oxygen species (ROS), which would cause a series of molecular and cellular damages, immune response, and eventually break down the tumor tissue. In recent years, catalytic nanosystems with unique structures and functions have emerged that can enhance X-PDT therapeutic effects via an immune response. The anti-cancer effect of X-PDT is closely related to the following factors: energy conversion efficiency of the material, the radiation dose of X-rays, quantum yield of the material, tumor resistance, and biocompatibility. Based on the latest research in this field and the classical theories of nanoscience, this paper systematically elucidates the current development of the X-PDT and related immunotherapy, and highlights its broad prospects in medical applications, discussing the connection between fundamental science and clinical translation.
Collapse
Affiliation(s)
- Gang Zhang
- Department of Physics, School of Science, Tianjin Chengjian University, Tianjin 300384, China.
| | - Meili Guo
- Department of Physics, School of Science, Tianjin Chengjian University, Tianjin 300384, China.
| | - Huizhen Ma
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China.
| | - Junying Wang
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Xiao-Dong Zhang
- Department of Physics and Tianjin Key Laboratory of Low Dimensional Materials Physics and Preparing Technology, School of Sciences, Tianjin University, Tianjin 300350, China. .,Tianjin Key Laboratory of Brain Science and Neural Engineering, Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| |
Collapse
|
15
|
He L, Yu X, Li W. Recent Progress and Trends in X-ray-Induced Photodynamic Therapy with Low Radiation Doses. ACS NANO 2022; 16:19691-19721. [PMID: 36378555 DOI: 10.1021/acsnano.2c07286] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The prominence of photodynamic therapy (PDT) in treating superficial skin cancer inspires innovative solutions for its congenitally deficient shadow penetration of the visible-light excitation. X-ray-induced photodynamic therapy (X-PDT) has been proven to be a successful technique in reforming the conventional PDT for deep-seated tumors by creatively utilizing penetrating X-rays as external excitation sources and has witnessed rapid developments over the past several years. Beyond the proof-of-concept demonstration, recent advances in X-PDT have exhibited a trend of minimizing X-ray radiation doses to quite low values. As such, scintillating materials used to bridge X-rays and photosensitizers play a significant role, as do diverse well-designed irradiation modes and smart strategies for improving the tumor microenvironment. Here in this review, we provide a comprehensive summary of recent achievements in X-PDT and highlight trending efforts using low doses of X-ray radiation. We first describe the concept of X-PDT and its relationships with radiodynamic therapy and radiotherapy and then dissect the mechanism of X-ray absorption and conversion by scintillating materials, reactive oxygen species evaluation for X-PDT, and radiation side effects and clinical concerns on X-ray radiation. Finally, we discuss a detailed overview of recent progress regarding low-dose X-PDT and present perspectives on possible clinical translation. It is expected that the pursuit of low-dose X-PDT will facilitate significant breakthroughs, both fundamentally and clinically, for effective deep-seated cancer treatment in the near future.
Collapse
|
16
|
Advanced techniques for performing photodynamic therapy in deep-seated tissues. Biomaterials 2022; 291:121875. [PMID: 36335717 DOI: 10.1016/j.biomaterials.2022.121875] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/07/2022] [Accepted: 10/23/2022] [Indexed: 11/23/2022]
Abstract
Photodynamic therapy (PDT) is a promising localized cancer treatment modality. It has been used successfully to treat a range of dermatological conditions with comparable efficacy to conventional treatments. However, some drawbacks limit the clinical utility of PDT in treating deep-seated tumors. Notably, the penetration limitation of UV and visible light, commonly applied to activate photosensitizers, makes PDT incompetent in treating deep-seated tumors. Development in light delivery technologies, especially fiber optics, led to improved clinical strategies for accessing deep tissues for irradiation. However, PDT efficacy issues remained partly due to light penetration limitations. In this review, we first summarized the current PDT applications for deep-seated tumor treatment. Then, the most recent progress in advanced techniques to overcome the light penetration limitation in PDT, including using functional nanomaterials that can either self-illuminate or be activated by near-infrared (NIR) light and X-rays as transducers, and implantable light delivery devices were discussed. Finally, current challenges and future opportunities of these technologies were discussed, which we hope may inspire the development of more effective techniques to enhance PDT efficacy against deep-seated tumors.
Collapse
|
17
|
Viswanath D, Won YY. Combining Radiotherapy (RT) and Photodynamic Therapy (PDT): Clinical Studies on Conventional RT-PDT Approaches and Novel Nanoparticle-Based RT-PDT Approaches under Preclinical Evaluation. ACS Biomater Sci Eng 2022; 8:3644-3658. [PMID: 36000986 DOI: 10.1021/acsbiomaterials.2c00287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Radiotherapy (RT) is the primary standard of care for many locally advanced cancers. Often times, however, the efficacy of RT is limited due to radio-resistance that cancer cells develop. Photodynamic therapy (PDT) has gained importance as an alternative local therapy. Because its mechanism involves minimal acquired resistance, PDT is a useful adjunct to RT. This review discusses recent advances in combining RT with PDT for cancer treatment. In the first part of this review, we will discuss clinical trials on RT + PDT combination therapies. All these approaches suffer from the same inherent limitations as any current PDT methods; (i) visible light has a short penetration depth in human tissue (<∼10 mm), and (ii) it is difficult to illuminate the entire tumor homogeneously by external/interstitial laser irradiation. To address these limitations, scintillating nanoparticle-mediated RT-PDT approaches have been explored in which nanoparticles convert X-rays (RT) into visible light (PDT); high-energy X-rays can reach deep into the body to irradiate cancers uniformly and precisely. The second part of this review will discuss recent efforts in developing and applying nanoparticles for RT-PDT applications.
Collapse
Affiliation(s)
- Dhushyanth Viswanath
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - You-Yeon Won
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States.,Purdue University Center for Cancer Research, West Lafayette, Indiana 47906, United States
| |
Collapse
|
18
|
Dhaini B, Wagner L, Moinard M, Daouk J, Arnoux P, Schohn H, Schneller P, Acherar S, Hamieh T, Frochot C. Importance of Rose Bengal Loaded with Nanoparticles for Anti-Cancer Photodynamic Therapy. Pharmaceuticals (Basel) 2022; 15:ph15091093. [PMID: 36145315 PMCID: PMC9504923 DOI: 10.3390/ph15091093] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/09/2022] [Accepted: 08/16/2022] [Indexed: 11/23/2022] Open
Abstract
Rose Bengal (RB) is a photosensitizer (PS) used in anti-cancer and anti-bacterial photodynamic therapy (PDT). The specific excitation of this PS allows the production of singlet oxygen and oxygen reactive species that kill bacteria and tumor cells. In this review, we summarize the history of the use of RB as a PS coupled by chemical or physical means to nanoparticles (NPs). The studies are divided into PDT and PDT excited by X-rays (X-PDT), and subdivided on the basis of NP type. On the basis of the papers examined, it can be noted that RB used as a PS shows remarkable cytotoxicity under the effect of light, and RB loaded onto NPs is an excellent candidate for nanomedical applications in PDT and X-PDT.
Collapse
Affiliation(s)
- Batoul Dhaini
- Reactions and Chemical Engineering Laboratory, Université de Lorraine, LRGP-CNRS, F-54000 Nancy, France
| | - Laurène Wagner
- Laboratory of Macromolecular Physical Chemistry, Université de Lorraine, LCPM-CNRS, F-54000 Nancy, France
| | - Morgane Moinard
- Reactions and Chemical Engineering Laboratory, Université de Lorraine, LRGP-CNRS, F-54000 Nancy, France
| | - Joël Daouk
- Department of Biology, Signals and Systems in Cancer and Neuroscience, Université de Lorraine, CRAN-CNRS, F-54000 Nancy, France
| | - Philippe Arnoux
- Reactions and Chemical Engineering Laboratory, Université de Lorraine, LRGP-CNRS, F-54000 Nancy, France
| | - Hervé Schohn
- Department of Biology, Signals and Systems in Cancer and Neuroscience, Université de Lorraine, CRAN-CNRS, F-54000 Nancy, France
| | - Perrine Schneller
- Department of Biology, Signals and Systems in Cancer and Neuroscience, Université de Lorraine, CRAN-CNRS, F-54000 Nancy, France
| | - Samir Acherar
- Laboratory of Macromolecular Physical Chemistry, Université de Lorraine, LCPM-CNRS, F-54000 Nancy, France
| | - Tayssir Hamieh
- Faculty of Science and Engineering, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Laboratory of Materials, Catalysis, Environment and Analytical Methods Laboratory (MCEMA), Faculty of Sciences, Lebanese University, Hadath 6573, Lebanon
| | - Céline Frochot
- Reactions and Chemical Engineering Laboratory, Université de Lorraine, LRGP-CNRS, F-54000 Nancy, France
- Correspondence:
| |
Collapse
|
19
|
Liu B, Liu C, Zhang X, Yao S, Wang Z, Liu Z, Song K, Li J. X-ray triggered pea-shaped LuAG:Mn/Ca nano-scintillators and their applications for photodynamic therapy. J Mater Chem B 2022; 10:6380-6391. [PMID: 35968697 DOI: 10.1039/d2tb01080a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Photodynamic therapy (PDT) is a new minimally invasive technology for disease diagnosis and treatment. However, the biological tissue attenuation of visible light renders the depth of its penetration in tissues quite modest, which significantly restricts its therapeutic applicability. Therefore, it is an essential but yet a difficult task to enhance the X-ray sensitization impact while concurrently limiting the tissue scattering by the rational design of novel biological vectors. Herein, a novel Lu3Al5O12:Mn/Ca-Ce6@SiO2 nanoparticle system (LAMCCS) based on a pea-shaped LuAG:Mn/Ca nano-scintillator (LAMC) activating photosensitizer agent (Ce6) was designed. Due to the high radiosensitization of LAMC nano-scintillators and efficient energy conversion efficiency between LAMC and Ce6, more singlet oxygen (1O2) could be generated to efficiently damage DNA fragments and reveal a good effect of inhibiting the long-term proliferation of tumor cells in vitro. Significantly, synergistic therapy with PDT/radiotherapy (RT) and from LAMCCS nanocomposites may still maintain a high tumor growth inhibition rate of 72% than single RT of 10% in vivo. Owing to their excellent ability for X-ray sensitization and energy conversion, LAMCCS nanocomposites may have significant tumor growth suppression rates under lower X-ray dose irradiation due to their outstanding X-ray sensitization and energy conversion capabilities, which may open up a new avenue for the advancement of cancer therapy.
Collapse
Affiliation(s)
- Bin Liu
- School of Material Science and Engineering, University of Jinan, Jinan, China.
| | - Chang Liu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Division of Gynecologic Oncology, Qilu Hospital of Shandong University, Jinan, China.
| | - Xiaolei Zhang
- School of Material Science and Engineering, University of Jinan, Jinan, China.
| | - Shu Yao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Division of Gynecologic Oncology, Qilu Hospital of Shandong University, Jinan, China.
| | - Ziying Wang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Division of Gynecologic Oncology, Qilu Hospital of Shandong University, Jinan, China.
| | - Zongming Liu
- School of Material Science and Engineering, University of Jinan, Jinan, China.
| | - Kun Song
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Division of Gynecologic Oncology, Qilu Hospital of Shandong University, Jinan, China.
| | - Jinkai Li
- School of Material Science and Engineering, University of Jinan, Jinan, China.
- Infovision Optoelectronics(Kunshan)Co, Ltd, Kunshan, China
| |
Collapse
|
20
|
Secchi V, Monguzzi A, Villa I. Design Principles of Hybrid Nanomaterials for Radiotherapy Enhanced by Photodynamic Therapy. Int J Mol Sci 2022; 23:8736. [PMID: 35955867 PMCID: PMC9369190 DOI: 10.3390/ijms23158736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/28/2022] [Accepted: 08/03/2022] [Indexed: 11/24/2022] Open
Abstract
Radiation (RT) remains the most frequently used treatment against cancer. The main limitation of RT is its lack of specificity for cancer tissues and the limited maximum radiation dose that can be safely delivered without damaging the surrounding healthy tissues. A step forward in the development of better RT is achieved by coupling it with other treatments, such as photodynamic therapy (PDT). PDT is an anti-cancer therapy that relies on the light activation of non-toxic molecules-called photosensitizers-to generate ROS such as singlet oxygen. By conjugating photosensitizers to dense nanoscintillators in hybrid architectures, the PDT could be activated during RT, leading to cell death through an additional pathway with respect to the one activated by RT alone. Therefore, combining RT and PDT can lead to a synergistic enhancement of the overall efficacy of RT. However, the involvement of hybrids in combination with ionizing radiation is not trivial: the comprehension of the relationship among RT, scintillation emission of the nanoscintillator, and therapeutic effects of the locally excited photosensitizers is desirable to optimize the design of the hybrid nanoparticles for improved effects in radio-oncology. Here, we discuss the working principles of the PDT-activated RT methods, pointing out the guidelines for the development of effective coadjutants to be tested in clinics.
Collapse
Affiliation(s)
- Valeria Secchi
- Department of Materials Science, University of Milano-Bicocca, Via R. Cozzi 55, 20125 Milan, Italy
- NANOMIB, Center for Biomedical Nanomedicine, University of Milano-Bicocca, P.zza Ateneo Nuovo 1, 20126 Milan, Italy
| | - Angelo Monguzzi
- Department of Materials Science, University of Milano-Bicocca, Via R. Cozzi 55, 20125 Milan, Italy
- NANOMIB, Center for Biomedical Nanomedicine, University of Milano-Bicocca, P.zza Ateneo Nuovo 1, 20126 Milan, Italy
| | - Irene Villa
- Institute of Physics of the Czech Academy of Sciences, FZU, Cukrovarnická 10/112, 16200 Prague, Czech Republic
| |
Collapse
|
21
|
Koohi Moftakhari Esfahani M, Alavi SE, Cabot PJ, Islam N, Izake EL. Application of Mesoporous Silica Nanoparticles in Cancer Therapy and Delivery of Repurposed Anthelmintics for Cancer Therapy. Pharmaceutics 2022; 14:pharmaceutics14081579. [PMID: 36015204 PMCID: PMC9415106 DOI: 10.3390/pharmaceutics14081579] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022] Open
Abstract
This review focuses on the biomedical application of mesoporous silica nanoparticles (MSNs), mainly focusing on the therapeutic application of MSNs for cancer treatment and specifically on overcoming the challenges of currently available anthelmintics (e.g., low water solubility) as repurposed drugs for cancer treatment. MSNs, due to their promising features, such as tunable pore size and volume, ability to control the drug release, and ability to convert the crystalline state of drugs to an amorphous state, are appropriate carriers for drug delivery with the improved solubility of hydrophobic drugs. The biomedical applications of MSNs can be further improved by the development of MSN-based multimodal anticancer therapeutics (e.g., photosensitizer-, photothermal-, and chemotherapeutics-modified MSNs) and chemical modifications, such as poly ethyleneglycol (PEG)ylation. In this review, various applications of MSNs (photodynamic and sonodynamic therapies, chemotherapy, radiation therapy, gene therapy, immunotherapy) and, in particular, as the carrier of anthelmintics for cancer therapy have been discussed. Additionally, the issues related to the safety of these nanoparticles have been deeply discussed. According to the findings of this literature review, the applications of MSN nanosystems for cancer therapy are a promising approach to improving the efficacy of the diagnostic and chemotherapeutic agents. Moreover, the MSN systems seem to be an efficient strategy to further help to decrease treatment costs by reducing the drug dose.
Collapse
Affiliation(s)
- Maedeh Koohi Moftakhari Esfahani
- School of Chemistry and Physics, Faculty of Science, Queensland University of Technology (QUT), 2 George Street, Brisbane, QLD 4000, Australia;
- Centre for Materials Science, Queensland University of Technology (QUT), 2 George Street, Brisbane, QLD 4000, Australia
| | - Seyed Ebrahim Alavi
- School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4215, Australia;
| | - Peter J. Cabot
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4102, Australia;
| | - Nazrul Islam
- School of Clinical Sciences, Faculty of Health, Queensland University of Technology, 2 George Street, Brisbane, QLD 4000, Australia;
- Centre for Immunology and Infection Control (CIIC), Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Emad L. Izake
- School of Chemistry and Physics, Faculty of Science, Queensland University of Technology (QUT), 2 George Street, Brisbane, QLD 4000, Australia;
- Centre for Materials Science, Queensland University of Technology (QUT), 2 George Street, Brisbane, QLD 4000, Australia
- Correspondence: ; Tel.: +61-7-3138-2501
| |
Collapse
|
22
|
Wang Y, Zhang H, Liu Y, Younis MH, Cai W, Bu W. Catalytic radiosensitization: Insights from materials physicochemistry. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2022; 57:262-278. [PMID: 36425004 PMCID: PMC9681018 DOI: 10.1016/j.mattod.2022.05.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Radiotherapy is indispensable in clinical cancer treatment, but because both tumor and normal tissues have similar sensitivity to X-rays, their clinical curative effect is intrinsically limited. Advanced nanomaterials and nanotechnologies have been developed for radiotherapy sensitization, typically employing high atomic number (high-Z) materials to enhance the energy deposition of X-rays in tumor tissues, but the efficiency is largely limited by the toxicity of heavy metals. A new and promising approach for radiosensitization is catalytic radiosensitization, which takes advantage of the catalytic activity of nanomaterials triggered by radiation. The efficiency of catalytic radiosensitization can be greatly enhanced by electron modulation and energy conversion of nanocatalysts upon X-ray irradiation, further enhancing the clinical curative effect. In this review, we highlight the challenges and opportunities in cancer radiosensitization, discuss novel approaches to catalytic radiosensitization, and finally describe the development of catalytic radiosensitization based on an in-depth understanding of radio-nano interactions and catalysis-biological interactions.
Collapse
Affiliation(s)
- Ya Wang
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, PR China
| | - Huilin Zhang
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, PR China
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Yanyan Liu
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, PR China
| | - Muhsin H. Younis
- Department of Radiology and Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Weibo Cai
- Department of Radiology and Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Wenbo Bu
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200433, PR China
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| |
Collapse
|
23
|
Yang J, Huang L, Qian K. Nanomaterials-assisted metabolic analysis toward in vitro diagnostics. EXPLORATION (BEIJING, CHINA) 2022; 2:20210222. [PMID: 37323704 PMCID: PMC10191060 DOI: 10.1002/exp.20210222] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/08/2022] [Indexed: 06/15/2023]
Abstract
In vitro diagnostics (IVD) has played an indispensable role in healthcare system by providing necessary information to indicate disease condition and guide therapeutic decision. Metabolic analysis can be the primary choice to facilitate the IVD since it characterizes the downstream metabolites and offers real-time feedback of the human body. Nanomaterials with well-designed composition and nanostructure have been developed for the construction of high-performance detection platforms toward metabolic analysis. Herein, we summarize the recent progress of nanomaterials-assisted metabolic analysis and the related applications in IVD. We first introduce the important role that nanomaterials play in metabolic analysis when coupled with different detection platforms, including electrochemical sensors, optical spectrometry, and mass spectrometry. We further highlight the nanomaterials-assisted metabolic analysis toward IVD applications, from the perspectives of both the targeted biomarker quantitation and untargeted fingerprint extraction. This review provides fundamental insights into the function of nanomaterials in metabolic analysis, thus facilitating the design of next-generation diagnostic devices in clinical practice.
Collapse
Affiliation(s)
- Jing Yang
- State Key Laboratory for Oncogenes and Related Genes, School of Biomedical Engineering, Institute of Medical Robotics and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghaiChina
- Department of Obstetrics and Gynecology, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Lin Huang
- Country Department of Clinical Laboratory MedicineShanghai Chest HospitalShanghai Jiao Tong UniversityShanghaiChina
| | - Kun Qian
- State Key Laboratory for Oncogenes and Related Genes, School of Biomedical Engineering, Institute of Medical Robotics and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghaiChina
- Department of Obstetrics and Gynecology, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
24
|
Chen L, Huang J, Li X, Huang M, Zeng S, Zheng J, Peng S, Li S. Progress of Nanomaterials in Photodynamic Therapy Against Tumor. Front Bioeng Biotechnol 2022; 10:920162. [PMID: 35711646 PMCID: PMC9194820 DOI: 10.3389/fbioe.2022.920162] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/02/2022] [Indexed: 12/19/2022] Open
Abstract
Photodynamic therapy (PDT) is an advanced therapeutic strategy with light-triggered, minimally invasive, high spatiotemporal selective and low systemic toxicity properties, which has been widely used in the clinical treatment of many solid tumors in recent years. Any strategies that improve the three elements of PDT (light, oxygen, and photosensitizers) can improve the efficacy of PDT. However, traditional PDT is confronted some challenges of poor solubility of photosensitizers and tumor suppressive microenvironment. To overcome the related obstacles of PDT, various strategies have been investigated in terms of improving photosensitizers (PSs) delivery, penetration of excitation light sources, and hypoxic tumor microenvironment. In addition, compared with a single treatment mode, the synergistic treatment of multiple treatment modalities such as photothermal therapy, chemotherapy, and radiation therapy can improve the efficacy of PDT. This review summarizes recent advances in nanomaterials, including metal nanoparticles, liposomes, hydrogels and polymers, to enhance the efficiency of PDT against malignant tumor.
Collapse
Affiliation(s)
- Lei Chen
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiahui Huang
- Department of Anesthesiology, Huizhou Central People’s Hospital, Huizhou, China
| | - Xiaotong Li
- Guangzhou Medical University, Guangzhou, China
| | | | | | - Jiayi Zheng
- Guangzhou Medical University, Guangzhou, China
| | - Shuyi Peng
- Guangzhou Medical University, Guangzhou, China
| | - Shiying Li
- Key Laboratory of Molecular Target and Clinical Pharmacology and The State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- *Correspondence: Shiying Li,
| |
Collapse
|
25
|
Hao X, Wu J, Xiang D, Yang Y. Recent Advance of Nanomaterial-Mediated Tumor Therapies in the Past Five Years. Front Pharmacol 2022; 13:846715. [PMID: 35250598 PMCID: PMC8896221 DOI: 10.3389/fphar.2022.846715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 01/31/2022] [Indexed: 12/07/2022] Open
Abstract
Cancer has posed a major threat to human life and health with a rapidly increasing number of patients. The complexity and refractory of tumors have brought great challenges to tumor treatment. In recent years, nanomaterials and nanotechnology have attracted more attention and greatly improved the efficiency of tumor therapies and significantly prolonged the survival period, whether for traditional tumor treatment methods such as radiotherapy, or emerging methods, such as phototherapy and immunotherapy, sonodynamic therapy, chemodynamic therapy and RNA interference therapeutics. Various monotherapies have obtained positive results, while combination therapies are further proposed to prevent incomplete eradication and recurrence of tumors, strengthen tumor killing efficacy with minimal side effects. In view of the complementary promotion effects between different therapies, it is vital to utilize nanomaterials as the link between monotherapies to achieve synergistic performance. Further development of nanomaterials with efficient tumor-killing effect and better biosafety is more in line with the needs of clinical treatment. In a word, the development of nanomaterials provides a promising way for tumor treatment, and here we will review the emerging nanomaterials towards radiotherapy, phototherapy and immunotherapy, and summarized the developed nanocarriers applied for the tumor combination therapies in the past 5 years, besides, the advances of some other novel therapies such as sonodynamic therapy, chemodynamic therapy, and RNA interference therapeutics have also been mentioned.
Collapse
Affiliation(s)
- Xinyan Hao
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Junyong Wu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - DaXiong Xiang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
| | - Yongyu Yang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Provincial Engineering Research Centre of Translational Medicine and Innovative Drug, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- *Correspondence: Yongyu Yang,
| |
Collapse
|
26
|
Li M, Bian X, Chen X, Fan N, Zou H, Bao Y, Zhou Y. Multifunctional liposome for photoacoustic/ultrasound imaging-guided chemo/photothermal retinoblastoma therapy. Drug Deliv 2022; 29:519-533. [PMID: 35156504 PMCID: PMC8863383 DOI: 10.1080/10717544.2022.2032876] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Retinoblastoma (RB) is a malignant intraocular neoplasm that occurs in children. Diagnosis and therapy are frequently delayed, often leading to metastasis, which necessitates effective imaging and treatment. In recent years, the use of nanoplatforms allowing both imaging and targeted treatment has attracted much attention. Herein, we report a novel nanoplatform folate-receptor (FR) targeted laser-activatable liposome termed FA-DOX-ICG-PFP@Lip, which is loaded with doxorubicin (DOX)/indocyanine green (ICG) and liquid perfluoropentane (PFP) for photoacoustic/ultrasound (PA/US) dual-modal imaging-guided chemo/photothermal RB therapy. The dual-modal imaging capability, photothermal conversion under laser irradiation, biocompatibility, and antitumor ability of these liposomes were appraised. The multifunctional liposome showed a good tumor targeting ability and was efficacious as a dual-modality contrast agent both in vivo and in vitro. When laser-irradiated, the liposome converted light energy to heat. This action caused immediate destruction of tumor cells, while simultaneously initiating PFP phase transformation to release DOX, resulting in both photothermal and chemotherapeutic antitumor effects. Notably, the FA-DOX-ICG-PFP@Lip showed good biocompatibility and no systemic toxicity was observed after laser irradiation in RB tumor-bearing mice. Hence, the FA-DOX-ICG-PFP@Lip shows great promise for dual-modal imaging-guided chemo/photothermal therapy, and may have significant value for diagnosing and treating RB.
Collapse
Affiliation(s)
- Meng Li
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China.,Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, PR China
| | - Xintong Bian
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China.,Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, PR China
| | - Xu Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China.,Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Sun Yat-sen University, Guangzhou, PR China
| | - Ningke Fan
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, PR China
| | - Hongmi Zou
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Yixi Bao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Yu Zhou
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| |
Collapse
|
27
|
Feng Y, Liu X, Li Q, Mei S, Wu K, Yuan J, Tu L, Que I, Tamburini F, Baldazzi F, Chan A, Cruz LJ, Zuo J, Yao C, Zhang H. A scintillating nanoplatform with upconversion function for the synergy of radiation and photodynamic therapies for deep tumors. JOURNAL OF MATERIALS CHEMISTRY. C 2022; 10:688-695. [PMID: 35127099 PMCID: PMC8740696 DOI: 10.1039/d1tc04930e] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/24/2021] [Indexed: 05/27/2023]
Abstract
Collaborative therapy is regarded as an effective approach in increasing the therapeutic efficacy of cancer. In this work, we have proposed and validated the concept of upconversion lumienscence image guided synergy of photodynamic therapy (PDT) and radiotherapy (RT) for deep cancer, via a specially designed nanoplatform integrating near infrared (NIR) light activated luminescence upconversion and X-ray induced scintillation. Upon NIR light irradiation, the nanoplatform emits highly monochromatic red light solely for imaging the targeted cancer cells without triggering therapy; however, when the irradiation turns to a low dose of X-rays, scintillation will occur which induces effectively the PDT destroying the cancer cells together with X-ray induced RT. The novel theranostic nanoplatform is constructed in such a way that the interactions between the upconversion core and the outmost scintillating shell are blocked effectively by an inert layer between them. This structural design not only enables a nearly perfect excitation energy delivery (∼100% at a spectral overlapping wavelength of ∼540 nm) from the outermost scintellating layer to the surface-anchored photosensitizers and so a maximum yield of radical oxygen species, but also achieves a strong NIR induced upconversion luminescence for imaging. Since PDT and RT attack different parts of a cancer cell, this synergy is more effective in destroying cancer than a single therapy, resulting in the reduction of the X-ray irradiation dosage. As a proof of principle, the theranostic effect is validated by in vitro and in vivo experiments, exhibiting the great potential of this sort of nanoplatform in deep cancer treatment.
Collapse
Affiliation(s)
- Yansong Feng
- State Key Laboratory of Explosion Science and Technology, School of Mechatronical Engineering, Beijing Institute of Technology 100081 Beijing China
- Van't Hoff Institute for Molecular Sciences, University of Amsterdam Science Park 904 1098 XH Amsterdam The Netherlands
| | - Xiaomeng Liu
- State Key Laboratory of Explosion Science and Technology, School of Mechatronical Engineering, Beijing Institute of Technology 100081 Beijing China
| | - Qiqing Li
- Van't Hoff Institute for Molecular Sciences, University of Amsterdam Science Park 904 1098 XH Amsterdam The Netherlands
| | - Shilin Mei
- State Key Laboratory of Explosion Science and Technology, School of Mechatronical Engineering, Beijing Institute of Technology 100081 Beijing China
| | - Kefan Wu
- Van't Hoff Institute for Molecular Sciences, University of Amsterdam Science Park 904 1098 XH Amsterdam The Netherlands
| | - Jun Yuan
- Van't Hoff Institute for Molecular Sciences, University of Amsterdam Science Park 904 1098 XH Amsterdam The Netherlands
| | - Langping Tu
- State Key Laboratory of Luminescence and Applications, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences 130033 Changchun China
| | - Ivo Que
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center 2333 ZA Leiden The Netherlands
| | | | - Fabio Baldazzi
- Percuros B.V. Zernikedreef 8 2333 CL Leiden The Netherlands
| | - Alan Chan
- Percuros B.V. Zernikedreef 8 2333 CL Leiden The Netherlands
| | - Luis J Cruz
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center 2333 ZA Leiden The Netherlands
| | - Jing Zuo
- Van't Hoff Institute for Molecular Sciences, University of Amsterdam Science Park 904 1098 XH Amsterdam The Netherlands
- Key Laboratory of Automobile Materials (Ministry of Education), College of Materials Science and Engineering, Jilin University 130025 Changchun China
| | - Changjiang Yao
- State Key Laboratory of Explosion Science and Technology, School of Mechatronical Engineering, Beijing Institute of Technology 100081 Beijing China
| | - Hong Zhang
- Van't Hoff Institute for Molecular Sciences, University of Amsterdam Science Park 904 1098 XH Amsterdam The Netherlands
| |
Collapse
|
28
|
Pan Y, Tang W, Fan W, Zhang J, Chen X. Development of nanotechnology-mediated precision radiotherapy for anti-metastasis and radioprotection. Chem Soc Rev 2022; 51:9759-9830. [DOI: 10.1039/d1cs01145f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Radiotherapy (RT), including external beam RT and internal radiation therapy, uses high-energy ionizing radiation to kill tumor cells.
Collapse
Affiliation(s)
- Yuanbo Pan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310009, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
| | - Wei Tang
- Departments of Pharmacy and Diagnostic Radiology, Nanomedicine Translational Research Program, Faculty of Science and Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117544, Singapore
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 210009, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, 310009, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, 310009, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|
29
|
An R, Liang Y, Du P, Lei P, Zhang H. Facile synthesis of rare earth-doped CeF 3 two-dimensional nanosheets and their application in ratiometric luminescence temperature sensing. CrystEngComm 2022. [DOI: 10.1039/d2ce00550f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Rare earth-doped CeF3 two-dimensional nanosheets have been successfully synthesized and their potential application as a ratiometric luminescent thermometer.
Collapse
Affiliation(s)
- Ran An
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| | - Yuan Liang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
- University of Science and Technology of China, Hefei 230026, China
- Ganjiang Innovation Academy, Chinese Academy of Sciences, Ganzhou, Jiangxi 341000, China
| | - Pengye Du
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
- University of Science and Technology of China, Hefei 230026, China
| | - Pengpeng Lei
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
| | - Hongjie Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, China
- University of Science and Technology of China, Hefei 230026, China
- Department of Chemistry, Tsinghua University, Beijing 100084, China
| |
Collapse
|
30
|
Xu W, Lei L, Wang Y, Liu E, Chen L, Xu S. Modulating electron population pathways for time-dependent dynamic multicolor displays. MATERIALS HORIZONS 2021; 8:3443-3448. [PMID: 34723303 DOI: 10.1039/d1mh01405f] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Multicolor luminescent nanoparticles (NPs) show several potential emerging applications. In this work, we provide a new route that integrates the afterglow and upconversion (UC) that originate in a single activator to achieve color variations without the modulation of any other parameters. The Er3+ ions in Na3HfF7:Yb/Er NPs exhibit bright green afterglow upon X-ray irradiation and single-band red UC under 980 nm laser excitation, which are attributed to the significantly different electron population pathways. The UC intensity is stable and the afterglow decreases gradually over time, thus the output color is clearly changed from green to red naturally via illuminating the pre-X-ray-irradiated NPs with a 980 nm laser. Furthermore, the fine emission profiles of Er3+, Ho3+ and Tm3+ are achieved upon X-ray irradiation. Our results develop a new approach for time-dependent dynamic color displays and a simple route to revealing the electronic fine structures of lanthanide activators at room temperature.
Collapse
Affiliation(s)
- Weixin Xu
- Institute of Optoelectronic Materials and Devices, Key Laboratory of Rare Earth Optoelectronic Materials and Devices of Zhejiang Province, College of Optical and Electronic Technology, China Jiliang University, Hangzhou 310018, P. R. China.
| | - Lei Lei
- Institute of Optoelectronic Materials and Devices, Key Laboratory of Rare Earth Optoelectronic Materials and Devices of Zhejiang Province, College of Optical and Electronic Technology, China Jiliang University, Hangzhou 310018, P. R. China.
| | - Yubin Wang
- Institute of Optoelectronic Materials and Devices, Key Laboratory of Rare Earth Optoelectronic Materials and Devices of Zhejiang Province, College of Optical and Electronic Technology, China Jiliang University, Hangzhou 310018, P. R. China.
| | - Enyang Liu
- Institute of Optoelectronic Materials and Devices, Key Laboratory of Rare Earth Optoelectronic Materials and Devices of Zhejiang Province, College of Optical and Electronic Technology, China Jiliang University, Hangzhou 310018, P. R. China.
| | - Liang Chen
- Institute of Optoelectronic Materials and Devices, Key Laboratory of Rare Earth Optoelectronic Materials and Devices of Zhejiang Province, College of Optical and Electronic Technology, China Jiliang University, Hangzhou 310018, P. R. China.
| | - Shiqing Xu
- Institute of Optoelectronic Materials and Devices, Key Laboratory of Rare Earth Optoelectronic Materials and Devices of Zhejiang Province, College of Optical and Electronic Technology, China Jiliang University, Hangzhou 310018, P. R. China.
| |
Collapse
|
31
|
Ma J, Zhu W, Lei L, Deng D, Hua Y, Yang YM, Xu S, Prasad PN. Highly Efficient NaGdF 4:Ce/Tb Nanoscintillator with Reduced Afterglow and Light Scattering for High-Resolution X-ray Imaging. ACS APPLIED MATERIALS & INTERFACES 2021; 13:44596-44603. [PMID: 34516086 DOI: 10.1021/acsami.1c14503] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Scintillation-based X-ray excited optical luminescence (XEOL) imaging shows great potential applications in the fields of industrial security inspection and medical diagnosis. It is still a great challenge to achieve scintillators simultaneously with low toxicity, high stability, strong XEOL intensity, and weak afterglow as well as simple device processibility with weak light scattering. Herein, we introduce ethylenediaminetetraacetate (EDTA)-capped NaGdF4:10Ce/18Tb nanoparticles (NPs) as a highly sensitive nanoscintillator, which meets all of the abovementioned challenges. These NPs show comparable XEOL intensity to the commercial CsI (Tl) single crystal in the green region. We propose a mechanism that involves a new electron-captured path by Ce3+ ions and the promotion of energy migration from a trap center to surface quenchers via a Gd3+ sublattice, which greatly reduces the population in traps to produce significant reduction of afterglow. Moreover, by employing an ultrathin transparent NaGdF4:10Ce/18Tb film (0.045 mm) as a nanoscintillator screen for XEOL imaging, a high spatial resolution of 18.6 lp mm-1 is realized owing to the greatly limited optical scattering, which is superior to the commercial CsI (TI) scintillator and most reported lead halide perovskites. We demonstrate that doping Ce3+ ions can greatly limit X-ray-activated afterglow, enabling to use an ultrathin transparent fluoride NP-based nanoscintillator screen for high-quality XEOL imaging of various objects such as an electronics chip and biological tissue.
Collapse
Affiliation(s)
- Jinjing Ma
- College of Optical and Electronic Technology, China Jiliang University, Hangzhou, Zhejiang 310018, China
| | - Wenjuan Zhu
- College of Optical Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310018, China
| | - Lei Lei
- College of Optical and Electronic Technology, China Jiliang University, Hangzhou, Zhejiang 310018, China
| | - Degang Deng
- College of Optical and Electronic Technology, China Jiliang University, Hangzhou, Zhejiang 310018, China
| | - Youjie Hua
- College of Optical and Electronic Technology, China Jiliang University, Hangzhou, Zhejiang 310018, China
| | - Yang Michael Yang
- College of Optical Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310018, China
| | - Shiqing Xu
- College of Optical and Electronic Technology, China Jiliang University, Hangzhou, Zhejiang 310018, China
| | - Paras N Prasad
- Institute for Lasers, Photonics, and Biophotonics and Department of Chemistry, University at Buffalo, State University of New York, Buffalo, New York 14260, United States
| |
Collapse
|
32
|
Jiang M, Deng Z, Zeng S, Hao J. Recent progress on lanthanide scintillators for soft X‐ray‐triggered bioimaging and deep‐tissue theranostics. VIEW 2021. [DOI: 10.1002/viw.20200122] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Affiliation(s)
- Mingyang Jiang
- Synergetic Innovation Center for Quantum Effects and Application Key Laboratory of Low‐dimensional Quantum Structures and Quantum Control of Ministry of Education Key Laboratory for Matter Microstructure and Function of Hunan Province School of Physics and Electronics Hunan Normal University Changsha P. R. China
| | - Zhiming Deng
- Synergetic Innovation Center for Quantum Effects and Application Key Laboratory of Low‐dimensional Quantum Structures and Quantum Control of Ministry of Education Key Laboratory for Matter Microstructure and Function of Hunan Province School of Physics and Electronics Hunan Normal University Changsha P. R. China
| | - Songjun Zeng
- Synergetic Innovation Center for Quantum Effects and Application Key Laboratory of Low‐dimensional Quantum Structures and Quantum Control of Ministry of Education Key Laboratory for Matter Microstructure and Function of Hunan Province School of Physics and Electronics Hunan Normal University Changsha P. R. China
| | - Jianhua Hao
- Department of Applied Physics The Hong Kong Polytechnic University Kowloon Hong Kong P. R. China
| |
Collapse
|
33
|
Jiang Z, He L, Yu X, Yang Z, Wu W, Wang X, Mao R, Cui D, Chen X, Li W. Antiangiogenesis Combined with Inhibition of the Hypoxia Pathway Facilitates Low-Dose, X-ray-Induced Photodynamic Therapy. ACS NANO 2021; 15:11112-11125. [PMID: 34170115 DOI: 10.1021/acsnano.1c01063] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
X-ray-induced photodynamic therapy (XPDT) is overwhelmingly superior in treating deep-seated cancers. However, limitations remain, owing to a combination of the poor scintillation performance of the nanoscintillator, low energy transfer efficiency of the therapeutic nanoplatform, and hypoxic environment presented in the tumor tissue. Collectively, these reduce the curative effect of XPDT. Here, we report a highly efficient, low-dose XPDT realized by systematic optimization from scintillation efficiency, nanoplatform structure, to therapeutic approach. We developed a biocompatible, codoped CaF2 nanoscintillator that emitted sufficiently green radioluminescence that was bright enough to be seen by the naked eye. Using dendrimers as a framework, we built a nanoplatform featuring a dual-core-satellite architecture, which enabled both procedurally and spatially separate dual-loading of therapeutic agents. This strategy allowed for the fabrication of a combined XPDT and antiangiogenic therapy, resulting in a therapeutic system capable of simultaneous tumor attacks. After exposure to ultralow dose radiation, XPDT resulted in marked tumor reduction while the antiangiogenic drug effectively blocked tumor vascularization exacerbated by XPDT-mediated hypoxia, rendering a pronounced synergy effect. This system also showed high biosafety, as the agents adopted had been used clinically and both Ca and F elements were widespread in the human body. Taken together, the findings presented here provided a reference for the construction of complex, multiloading architecture in coordination with structural complexity and functional diversification. This work provided a safer and more robust application of the combined XPDT and antiangiogenesis in future clinical treatment settings.
Collapse
Affiliation(s)
- Zhao Jiang
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Liangrui He
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Xujiang Yu
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Zhiwen Yang
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Weijie Wu
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Xiaoyan Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Rihua Mao
- Laboratory for Advanced Scintillation Materials & Performance, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 201800, PR China
| | - Daxiang Cui
- Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Key Laboratory of Thin Film and Microfabrication (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Xiaoyuan Chen
- Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, 117597 Singapore
| | - Wanwan Li
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, PR China
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, PR China
| |
Collapse
|
34
|
Kirsanova DY, Gadzhimagomedova ZM, Maksimov AY, Soldatov AV. Nanomaterials for Deep Tumor Treatment. Mini Rev Med Chem 2021; 21:677-688. [PMID: 33176645 DOI: 10.2174/1389557520666201111161705] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/25/2020] [Accepted: 08/20/2020] [Indexed: 11/22/2022]
Abstract
According to statistics, cancer is the second leading cause of death in the world. Thus, it is important to solve this medical and social problem by developing new effective methods for cancer treatment. An alternative to more well-known approaches, such as radiotherapy and chemotherapy, is photodynamic therapy (PDT), which is limited to the shallow tissue penetration (< 1 cm) of visible light. Since the PDT process can be initiated in deep tissues by X-ray irradiation (X-ray induced PDT, or XPDT), it has a great potential to treat tumors in internal organs. The article discusses the principles of therapies. The main focus is on various nanoparticles used with or without photosensitizers, which allow the conversion of X-ray irradiation into UV-visible light. Much attention is given to the synthesis of nanoparticles and analysis of their characteristics, such as size and spectral features. The results of in vitro and in vivo experiments are also discussed.
Collapse
Affiliation(s)
- Daria Yu Kirsanova
- The Smart Materials Research Institute, Southern Federal University, Sladkova 178/24, 344090, Rostov-on-Don, Russian Federation
| | - Zaira M Gadzhimagomedova
- The Smart Materials Research Institute, Southern Federal University, Sladkova 178/24, 344090, Rostov-on-Don, Russian Federation
| | - Aleksey Yu Maksimov
- National Medical Research Centre for Oncology, 14 liniya str. 63, 344037, Rostov-on-Don, Russian Federation
| | - Alexander V Soldatov
- The Smart Materials Research Institute, Southern Federal University, Sladkova 178/24, 344090, Rostov-on-Don, Russian Federation
| |
Collapse
|
35
|
Rastegari E, Hsiao YJ, Lai WY, Lai YH, Yang TC, Chen SJ, Huang PI, Chiou SH, Mou CY, Chien Y. An Update on Mesoporous Silica Nanoparticle Applications in Nanomedicine. Pharmaceutics 2021; 13:1067. [PMID: 34371758 PMCID: PMC8309088 DOI: 10.3390/pharmaceutics13071067] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 01/09/2023] Open
Abstract
The efficient and safe delivery of therapeutic drugs, proteins, and nucleic acids are essential for meaningful therapeutic benefits. The field of nanomedicine shows promising implications in the development of therapeutics by delivering diagnostic and therapeutic compounds. Nanomedicine development has led to significant advances in the design and engineering of nanocarrier systems with supra-molecular structures. Smart mesoporous silica nanoparticles (MSNs), with excellent biocompatibility, tunable physicochemical properties, and site-specific functionalization, offer efficient and high loading capacity as well as robust and targeted delivery of a variety of payloads in a controlled fashion. Such unique nanocarriers should have great potential for challenging biomedical applications, such as tissue engineering, bioimaging techniques, stem cell research, and cancer therapies. However, in vivo applications of these nanocarriers should be further validated before clinical translation. To this end, this review begins with a brief introduction of MSNs properties, targeted drug delivery, and controlled release with a particular emphasis on their most recent diagnostic and therapeutic applications.
Collapse
Grants
- MOST 108-2320-B-010 -019 -MY3; MOST 109-2327-B-010-007 Ministry of Science and Technology
- MOHW108-TDU-B-211-133001, MOHW109-TDU-B-211-114001 Ministry of Health and Welfare
- VN109-16 VGH, NTUH Joint Research Program
- VTA107-V1-5-1, VTA108-V1-5-3, VTA109-V1-4-1 VGH, TSGH, NDMC, AS Joint Research Program
- IBMS-CRC109-P04 AS Clinical Research Center
- the "Cancer Progression Research Center, National Yang-Ming University" from The Featured Areas Research Center Program within the framework of the Higher Education Sprout Project by the Ministry of Education (MOE) in Taiwan the "Cancer Progression Research Center, National Yang-Ming University" from The Featured Areas Research Center Program within the framework of the Higher Education Sprout Project by the Ministry of Education (MOE) in Taiwan
- and the Ministry of Education through the SPROUT Project- Center For Intelligent Drug Systems and Smart Bio-devices (IDS2B) of National Chiao Tung University and, Taiwan. and the Ministry of Education through the SPROUT Project- Center For Intelligent Drug Systems and Smart Bio-devices (IDS2B) of National Chiao Tung University and, Taiwan.
Collapse
Affiliation(s)
- Elham Rastegari
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (E.R.); (Y.-J.H.); (W.-Y.L.); (Y.-H.L.); (T.-C.Y.); (S.-J.C.)
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
| | - Yu-Jer Hsiao
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (E.R.); (Y.-J.H.); (W.-Y.L.); (Y.-H.L.); (T.-C.Y.); (S.-J.C.)
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
| | - Wei-Yi Lai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (E.R.); (Y.-J.H.); (W.-Y.L.); (Y.-H.L.); (T.-C.Y.); (S.-J.C.)
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
| | - Yun-Hsien Lai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (E.R.); (Y.-J.H.); (W.-Y.L.); (Y.-H.L.); (T.-C.Y.); (S.-J.C.)
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
| | - Tien-Chun Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (E.R.); (Y.-J.H.); (W.-Y.L.); (Y.-H.L.); (T.-C.Y.); (S.-J.C.)
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
| | - Shih-Jen Chen
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (E.R.); (Y.-J.H.); (W.-Y.L.); (Y.-H.L.); (T.-C.Y.); (S.-J.C.)
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Pin-I Huang
- Department of Oncology, Taipei Veterans General Hospital, Taipei Veterans General Hospital, Taipei 11217, Taiwan;
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (E.R.); (Y.-J.H.); (W.-Y.L.); (Y.-H.L.); (T.-C.Y.); (S.-J.C.)
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Chung-Yuan Mou
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Yueh Chien
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan; (E.R.); (Y.-J.H.); (W.-Y.L.); (Y.-H.L.); (T.-C.Y.); (S.-J.C.)
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei 11217, Taiwan
| |
Collapse
|
36
|
Zhong X, Wang X, Li J, Hu J, Cheng L, Yang X. ROS-based dynamic therapy synergy with modulating tumor cell-microenvironment mediated by inorganic nanomedicine. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.213828] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
37
|
Screening of metabolites in the treatment of liver cancer xenografts HepG2/ADR by psoralen-loaded lipid nanoparticles. Eur J Pharm Biopharm 2021; 165:337-344. [PMID: 34062256 DOI: 10.1016/j.ejpb.2021.05.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Our study aimed to find potential biomarkers for drug resistance in liver cancer cells using metabolomics and further to evaluate the potential of psoralen-loaded polymer lipid nanoparticles (PSO-PLNs) to reverse the resistance of cells to doxorubicin. METHODS We used LC-MS-based non-targeted metabolomics, also known as global metabolite profiling, to screen in serum and urine of mice engrafted with a liver cancer cell line sensitive (HepG2/S) or resistant to doxorubicin (HepG2/ADR) for differentially regulated metabolites. We subsequently quantified the abundance of these metabolites in serum and the urine of mice. The mice were engrafted with HepG2 cells resistant against doxorubicin and were treated with I) doxorubicin, II) a combination of doxorubicin and psoralen and III) a combination of doxorubicin and psoralen packed in polymer lipid nanoparticles. RESULTS Metabolites found to be differentially present in urine of mice engrafted with resistant HepG2 cells were: hippuric acid, hyaluronic acid, pantothenic acid, and betaine; retinoic acid and α-linolenic acid were found to be reduced in serum samples of mice with HepG2 cells resistant to doxorubicin. The targeted analysis showed that the degree of regression of metabolic markers in groups differed: treatment group 2 had stronger degree of regression than treatment group 1 and the negative control group had the smallest, which indicates that the PSO-PLNs have superior properties compared with other treatments. CONCLUSION Psoralen reverses drug resistance of liver cancer cells and its efficacy can be increased by encapsulation in polymer lipid nanoparticles.
Collapse
|
38
|
Zhang X, Fu Q, Duan H, Song J, Yang H. Janus Nanoparticles: From Fabrication to (Bio)Applications. ACS NANO 2021; 15:6147-6191. [PMID: 33739822 DOI: 10.1021/acsnano.1c01146] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Janus nanoparticles (JNPs) refer to the integration of two or more chemically discrepant composites into one structure system. Studies into JNPs have been of significant interest due to their interesting characteristics stemming from their asymmetric structures, which can integrate different functional properties and perform more synergetic functions simultaneously. Herein, we present recent progress of Janus particles, comprehensively detailing fabrication strategies and applications. First, the classification of JNPs is divided into three blocks, consisting of polymeric composites, inorganic composites, and hybrid polymeric/inorganic JNPs composites. Then, the fabrication strategies are alternately summarized, examining self-assembly strategy, phase separation strategy, seed-mediated polymerization, microfluidic preparation strategy, nucleation growth methods, and masking methods. Finally, various intriguing applications of JNPs are presented, including solid surfactants agents, micro/nanomotors, and biomedical applications such as biosensing, controlled drug delivery, bioimaging, cancer therapy, and combined theranostics. Furthermore, challenges and future works in this field are provided.
Collapse
Affiliation(s)
- Xuan Zhang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, P.R. China
| | - Qinrui Fu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, P.R. China
| | - Hongwei Duan
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore 637457
| | - Jibin Song
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, P.R. China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, P.R. China
| |
Collapse
|
39
|
Zuo M, Duan Q, Li C, Ge J, Wang Q, Li Z, Liu Z. A Versatile Strategy for Constructing Ratiometric Upconversion Luminescent Probe with Sensitized Emission of Energy Acceptor. Anal Chem 2021; 93:5635-5643. [PMID: 33749233 DOI: 10.1021/acs.analchem.1c00470] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
When fabricating ratiometric optical probes using lanthanide-doped upconversion nanoparticles (UCNPs), which are promising luminescent materials that have widely been utilized in biosensing and bioimaging as energy donors, it is still a challenge to obtain the emission signal of energy acceptors with reasons unclear so far. Herein, we reveal that the energy-transfer efficiency and brightness of UCNPs as well as the aggregation-caused quenching (ACQ) of energy accepting dyes are the main factors restricting the emission of energy acceptors, and we have circumvented this problem by modulating the structure of UCNPs and the assembly manner of the energy donor-acceptor pair. On this basis, a proof-of-concept ratiometric upconversion nanoprobe was constructed for hydrogen sulfide (H2S) detection with an elaborate dye Fl-1 as an energy acceptor. As the H2S concentration increased, the emission intensity of Fl-1 at 525 nm increased gradually, accompanied by a decrease of upconversion luminescence at 480 nm, thus providing a ratiometric signal of F480/F525 dependent on the H2S concentration. This probe was able to track H2S in living cells and zebrafish and visualize the H2S level of mice in physiological processes.
Collapse
Affiliation(s)
- Miaomiao Zuo
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Qian Duan
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Chenchen Li
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Juan Ge
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Qirong Wang
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Zhen Li
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Zhihong Liu
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| |
Collapse
|
40
|
Nsubuga A, Mandl GA, Capobianco JA. Investigating the reactive oxygen species production of Rose Bengal and Merocyanine 540-loaded radioluminescent nanoparticles. NANOSCALE ADVANCES 2021; 3:1375-1381. [PMID: 36132856 PMCID: PMC9417208 DOI: 10.1039/d0na00964d] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/17/2021] [Indexed: 06/16/2023]
Abstract
Radioluminescent nanomaterials have garnered significant attention in the past decade due to their potential to perform X-ray mediated photodynamic therapy (X-PDT). Many of these materials are assumed to produce singlet oxygen based on a single assay. Herein we demonstrate that multiple assays are required to confidently determine whether singlet oxygen or other reactive oxygen species are being produced through type I or type II PDT mechanisms. Rose Bengal and Merocyanine 540 photosensitizers were loaded into mesoporous silica-coated NaLuF4:Dy3+,Gd3+ nanoparticles and the combination of ABDA, DPBF, and NaN3 assays along with electron paramagnetic resonance were employed to determine that superoxide and hydroxyl radicals were exclusively produced from this system under X-ray excitation. Knowledge of the correct PDT mechanism is crucial for informing what types of disease may be best suited for treatment using PDT nanosystems.
Collapse
Affiliation(s)
- Anne Nsubuga
- Department of Chemistry and Biochemistry, Centre for NanoScience Research, Concordia University 7141 Rue Sherbrooke Ouest Montreal QC H4B 1R6 Canada
| | - Gabrielle A Mandl
- Department of Chemistry and Biochemistry, Centre for NanoScience Research, Concordia University 7141 Rue Sherbrooke Ouest Montreal QC H4B 1R6 Canada
| | - John A Capobianco
- Department of Chemistry and Biochemistry, Centre for NanoScience Research, Concordia University 7141 Rue Sherbrooke Ouest Montreal QC H4B 1R6 Canada
| |
Collapse
|
41
|
Hu H, Feng W, Qian X, Yu L, Chen Y, Li Y. Emerging Nanomedicine-Enabled/Enhanced Nanodynamic Therapies beyond Traditional Photodynamics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2005062. [PMID: 33565157 DOI: 10.1002/adma.202005062] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/25/2020] [Indexed: 05/18/2023]
Abstract
The rapid knowledge growth of nanomedicine and nanobiotechnology enables and promotes the emergence of distinctive disease-specific therapeutic modalities, among which nanomedicine-enabled/augmented nanodynamic therapy (NDT), as triggered by either exogenous or endogenous activators on nanosensitizers, can generate reactive radicals for accomplishing efficient disease nanotherapies with mitigated side effects and endowed disease specificity. As one of the most representative modalities of NDT, traditional light-activated photodynamics suffers from the critical and unsurmountable issues of the low tissue-penetration depth of light and the phototoxicity of the photosensitizers. To overcome these obstacles, versatile nanomedicine-enabled/augmented NDTs have been explored for satisfying varied biomedical applications, which strongly depend on the physicochemical properties of the involved nanomedicines and nanosensitizers. These distinctive NDTs refer to sonodynamic therapy (SDT), thermodynamic therapy (TDT), electrodynamic therapy (EDT), piezoelectric dynamic therapy (PZDT), pyroelectric dynamic therapy (PEDT), radiodynamic therapy (RDT), and chemodynamic therapy (CDT). Herein, the critical roles, functions, and biological effects of nanomedicine (e.g., sonosensitizing, photothermal-converting, electronic, piezoelectric, pyroelectric, radiation-sensitizing, and catalytic properties) for enabling the therapeutic procedure of NDTs, are highlighted and discussed, along with the underlying therapeutic principle and optimization strategy for augmenting disease-therapeutic efficacy and biosafety. The present challenges and critical issues on the clinical translations of NDTs are also discussed and clarified.
Collapse
Affiliation(s)
- Hui Hu
- Medmaterial Research Center, Jiangsu University Affiliated People's Hospital, Zhenjiang, 212002, P. R. China
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Wei Feng
- School of Life Sciences, Shanghai University, Shanghai, 2000444, P. R. China
| | - Xiaoqin Qian
- Medmaterial Research Center, Jiangsu University Affiliated People's Hospital, Zhenjiang, 212002, P. R. China
| | - Luodan Yu
- School of Life Sciences, Shanghai University, Shanghai, 2000444, P. R. China
| | - Yu Chen
- School of Life Sciences, Shanghai University, Shanghai, 2000444, P. R. China
- State Key Laboratory of High Performance Ceramic and Superfine, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Yuehua Li
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| |
Collapse
|
42
|
Liu X, Yang S, Li Y, Wang B, Guo J, Ma X. Mesoporous Nanostructures Encapsulated with Metallic Nanodots for Smart SERS Sensing. ACS APPLIED MATERIALS & INTERFACES 2021; 13:186-195. [PMID: 33351580 DOI: 10.1021/acsami.0c17316] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
In virtue of uniform mesopores and core-shell nanoarchitectures, metallic nanodot-encapsulated hollow mesoporous nanostructures have shown promising potential in various applications. However, their fabrication with versatile tunability of the encapsulated metallic content has been a challenge. Herein, we have prepared metallic nanodot-encapsulated hollow mesoporous silica nanoparticles (M-HMSNPs) with adjustable inner metallic components. The sacrificial template of polystyrene (PS) nanoparticles precoated with metals (Au/Ag/Pt) is fully wrapped with mesoporous silica (mSiO2). The metallic nanodots are formed during the template removal process by calcination. The type and content of the encapsulated nanodots can be readily and precisely controlled by the initially deposited metallic layers. We demonstrate the application of the gold (Au) nanodot-loaded HMSNPs (denoted Au-HMSNPs) as smart surface-enhanced Raman spectroscopy (SERS) probes, which can screen between big molecules and small analytes. With the aid of a Raman reporter, the SERS probe can successfully quantify H2O2, which is used to distinguish cancer cells in vitro. Further integrated with enzymes, the SERS chips of specificity are prepared and used to detect corresponding substrates of glucose and uric acid, responsively. Besides SERS sensing, the current strategy can inspire future development of many other M-HMSNPs for various applications such as catalysis, energy storage, theranostics, etc.
Collapse
Affiliation(s)
- Xiaojia Liu
- Flexible Printed Electronic Technology Center and School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, China
- Shenzhen Bay Laboratory, No. 9 Duxue Road, Shenzhen 518055, China
| | - Shikun Yang
- Flexible Printed Electronic Technology Center and School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, China
- Shenzhen Bay Laboratory, No. 9 Duxue Road, Shenzhen 518055, China
| | - Yang Li
- School of Materials Engineering, Shanghai University of Engineering Science, Shanghai, 201620, China
| | - Bo Wang
- School of Materials Engineering, Shanghai University of Engineering Science, Shanghai, 201620, China
| | - Jinhong Guo
- School of Communication and Information Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Xing Ma
- Flexible Printed Electronic Technology Center and School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, China
- Shenzhen Bay Laboratory, No. 9 Duxue Road, Shenzhen 518055, China
| |
Collapse
|
43
|
Li WP, Yen CJ, Wu BS, Wong TW. Recent Advances in Photodynamic Therapy for Deep-Seated Tumors with the Aid of Nanomedicine. Biomedicines 2021; 9:69. [PMID: 33445690 PMCID: PMC7828119 DOI: 10.3390/biomedicines9010069] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 12/14/2022] Open
Abstract
Photodynamic therapy (PDT) works through photoactivation of a specific photosensitizer (PS) in a tumor in the presence of oxygen. PDT is widely applied in oncology to treat various cancers as it has a minimally invasive procedure and high selectivity, does not interfere with other treatments, and can be repeated as needed. A large amount of reactive oxygen species (ROS) and singlet oxygen is generated in a cancer cell during PDT, which destroys the tumor effectively. However, the efficacy of PDT in treating a deep-seated tumor is limited due to three main reasons: Limited light penetration depth, low oxygen concentration in the hypoxic core, and poor PS accumulation inside a tumor. Thus, PDT treatments are only approved for superficial and thin tumors. With the advancement of nanotechnology, PDT to treat deep-seated or thick tumors is becoming a reachable goal. In this review, we provide an update on the strategies for improving PDT with nanomedicine using different sophisticated-design nanoparticles, including two-photon excitation, X-ray activation, targeting tumor cells with surface modification, alteration of tumor cell metabolism pathways, release of therapeutic gases, improvement of tumor hypoxia, and stimulation of host immunity. We focus on the difficult-to-treat pancreatic cancer as a model to demonstrate the influence of advanced nanomedicine in PDT. A bright future of PDT application in the treatment of deep-seated tumors is expected.
Collapse
Affiliation(s)
- Wei-Peng Li
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chia-Jui Yen
- Division of Hematology and Oncology, Department of Internal Medicine, Graduate Institute of Clinical Medicine, National Cheng Kung University Hospital, Tainan 704, Taiwan;
| | - Bo-Sheng Wu
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Tak-Wah Wong
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan 701, Taiwan
| |
Collapse
|
44
|
Ahmad F, Abubshait SA, Abubshait HA. Untargeted metabolomics for Achilles heel of engineered nanomaterials' risk assessment. CHEMOSPHERE 2021; 262:128058. [PMID: 33182140 DOI: 10.1016/j.chemosphere.2020.128058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 06/11/2023]
Abstract
Owing to the superlative properties, engineered nanomaterials (ENM) are being used in food, cosmetics, medicine, and electronics. Therefore, exogenous ENM can be housed into humans through a multitude of exposure routes, leading to compromise of the biomolecules' functionalities through structural deformations, and even at the metabolic level. Consequently, it is of great importance to understand the perturbations introduced at the metabolic level for the timely risk assessment (RA) of ENM. Current technological advancements in metabolomics empower us to visualize the metabolic dysregulations in biological cells, tissues, and living objects, instigated by the ENM. Given the fact, we propose multitiered untargeted metabolomics for the risk assessment of ENM. We propose largely validated experimental design principles that enable the well-organized and authentic identification of metabolic dysregulation connected with a newly engineered nanomaterial. Our scheme could participate in the enhanced transparency of the RA course of rapidly emerging ENM.
Collapse
Affiliation(s)
- Farooq Ahmad
- School of Material Science and Engineering, Nanjing University, Jiangsu, China.
| | - Samar A Abubshait
- Department of Chemistry, College of Science, Imam Abdulrahman Bin Faisal University, 31441, Dammam, Saudi Arabia; Basic and Applied Scientific Research Center, Imam Abdulrahman Bin Faisal University, 31441, Dammam, Saudi Arabia
| | - Haya A Abubshait
- Basic Sciences Department, Deanship of Preparatory Year and Supporting Studies, Imam Adulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
45
|
Dong L, Li W, Sun L, Yu L, Chen Y, Hong G. Energy-converting biomaterials for cancer therapy: Category, efficiency, and biosafety. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 13:e1663. [PMID: 32808464 DOI: 10.1002/wnan.1663] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/08/2020] [Accepted: 07/10/2020] [Indexed: 12/24/2022]
Abstract
Energy-converting biomaterials (ECBs)-mediated cancer-therapeutic modalities have been extensively explored, which have achieved remarkable benefits to overwhelm the obstacles of traditional cancer-treatment modalities. Energy-driven cancer-therapeutic modalities feature their distinctive merits, including noninvasiveness, low mammalian toxicity, adequate therapeutic outcome, and optimistical synergistic therapeutics. In this advanced review, the prevailing mainstream ECBs can be divided into two sections: Reactive oxygen species (ROS)-associated energy-converting biomaterials (ROS-ECBs) and hyperthermia-related energy-converting biomaterials (H-ECBs). On the one hand, ROS-ECBs can transfer exogenous or endogenous energy (such as light, radiation, ultrasound, or chemical) to generate and release highly toxic ROS for inducing tumor cell apoptosis/necrosis, including photo-driven ROS-ECBs for photodynamic therapy, radiation-driven ROS-ECBs for radiotherapy, ultrasound-driven ROS-ECBs for sonodynamic therapy, and chemical-driven ROS-ECBs for chemodynamic therapy. On the other hand, H-ECBs could translate the external energy (such as light and magnetic) into heat for killing tumor cells, including photo-converted H-ECBs for photothermal therapy and magnetic-converted H-ECBs for magnetic hyperthermia therapy. Additionally, the biosafety issues of ECBs are expounded preliminarily, guaranteeing the ever-stringent requirements of clinical translation. Finally, we discussed the prospects and facing challenges for constructing the new-generation ECBs for establishing intriguing energy-driven cancer-therapeutic modalities. This article is categorized under: Nanotechnology Approaches to Biology >Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Lile Dong
- Department of Radiology, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Wenjuan Li
- Department of Radiology, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Lining Sun
- Research Center of Nano Science and Technology, College of Sciences, Shanghai University, Shanghai, China
| | - Luodan Yu
- School of Life Sciences, Shanghai University, Shanghai, China
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| | - Yu Chen
- School of Life Sciences, Shanghai University, Shanghai, China
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| | - Guobin Hong
- Department of Radiology, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| |
Collapse
|
46
|
Kuang Y, Zhang Y, Zhao Y, Cao Y, Zhang Y, Chong Y, Pei R. Dual-Stimuli-Responsive Multifunctional Gd 2Hf 2O 7 Nanoparticles for MRI-Guided Combined Chemo-/Photothermal-/Radiotherapy of Resistant Tumors. ACS APPLIED MATERIALS & INTERFACES 2020; 12:35928-35939. [PMID: 32686939 DOI: 10.1021/acsami.0c09422] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The design and synthesis of a novel generation of a nanoscaled platform with imaging-guided therapy remain a real challenge. It can not only improve the imaging sensitivity of tumor tissues for guiding all kinds of treatments but also reduce the harm for healthy tissues. Here, polydopamine (PDA), polyethylene glycol (PEG), and c(RGDyK) peptide (RGD)-modified and cisplatin-loaded Gd2Hf2O7 nanoparticles (Gd2Hf2O7@PDA@PEG-Pt-RGD NPs) are designed for magnetic resonance imaging (MRI)-guided combined chemo-/photothermal-/radiotherapy of resistant tumors. The as-prepared NPs display high relaxivity (r1 = 38.28 mM-1 s-1) as an MRI contrast agent because of their ultrasmall size and surface modification with polyacrylic acid and PDA. Gd2Hf2O7@PDA@PEG-Pt-RGD NPs exhibit pH and NIR dual-stimuli responsiveness for cisplatin release. Based on competent NIR absorption and high X-ray attenuation efficiency, Gd2Hf2O7@PDA@PEG-Pt-RGD NPs show potential photothermal effect by exposing to an 808 nm NIR laser and significantly improve the generation of reactive oxygen species after X-ray radiation. Combined chemo-/photothermal-/radiotherapy can effectively treat the resistant A549R cells, providing the enhanced therapeutic efficiency to cancer tissues and the reduced side effect to healthy tissues. Furthermore, Gd2Hf2O7@PDA@PEG-Pt-RGD NPs present no obvious toxicity during the treatment, which demonstrates the potential as an efficient MRI-guided combined chemo-/photothermal-/radiotherapy nanoplatform for drug-resistant tumors.
Collapse
Affiliation(s)
- Ye Kuang
- Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Fujian Medical University, Fuzhou 350004, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Ye Zhang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yuewu Zhao
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yi Cao
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yajie Zhang
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yu Chong
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Renjun Pei
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| |
Collapse
|
47
|
Yan K, Zhang Y, Mu C, Xu Q, Jing X, Wang D, Dang D, Meng L, Ma J. Versatile Nanoplatforms with enhanced Photodynamic Therapy: Designs and Applications. Theranostics 2020; 10:7287-7318. [PMID: 32641993 PMCID: PMC7330854 DOI: 10.7150/thno.46288] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/20/2020] [Indexed: 12/15/2022] Open
Abstract
As an emerging antitumor strategy, photodynamic therapy (PDT) has attracted intensive attention for the treatment of various malignant tumors owing to its noninvasive nature and high spatial selectivity in recent years. However, the therapeutic effect is unsatisfactory on some occasions due to the presence of some unfavorable factors including nonspecific accumulation of PS towards malignant tissues, the lack of endogenous oxygen in tumors, as well as the limited light penetration depth, further hampering practical application. To circumvent these limitations and improve real utilization efficiency, various enhanced strategies have been developed and explored during the past years. In this review, we give an overview of the state-of-the-art advances progress on versatile nanoplatforms for enhanced PDT considering the enhancement from targeting or responsive, chemical and physical effect. Specifically, these effects mainly include organelle-targeting function, tumor microenvironment responsive release photosensitizers (PS), self-sufficient O2 (affinity oxygen and generating oxygen), photocatalytic water splitting, X-rays light stimulate, surface plasmon resonance enhancement, and the improvement by resonance energy transfer. When utilizing these strategies to improve the therapeutic effect, the advantages and limitations are addressed. Finally, the challenges and prospective will be discussed and demonstrated for the future development of advanced PDT with enhanced efficacy.
Collapse
Affiliation(s)
- Kai Yan
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
- School of Science, Xi'an Key Laboratory of Sustainable Energy Material Chemistry, MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Materials, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Yabin Zhang
- Key Laboratory of Testing Technology for Manufacturing Process of Ministry of Education, Southwest University of Science and Technology, Mianyang 621010, P. R. China
- Institute of Textiles & Clothing, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Chenglong Mu
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Qunna Xu
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Xunan Jing
- School of Science, Xi'an Key Laboratory of Sustainable Energy Material Chemistry, MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Materials, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Daquan Wang
- School of Science, Xi'an Key Laboratory of Sustainable Energy Material Chemistry, MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Materials, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Dongfeng Dang
- School of Science, Xi'an Key Laboratory of Sustainable Energy Material Chemistry, MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Materials, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Lingjie Meng
- School of Science, Xi'an Key Laboratory of Sustainable Energy Material Chemistry, MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Materials, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Jianzhong Ma
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| |
Collapse
|
48
|
Gadzhimagomedova Z, Zolotukhin P, Kit O, Kirsanova D, Soldatov A. Nanocomposites for X-Ray Photodynamic Therapy. Int J Mol Sci 2020; 21:ijms21114004. [PMID: 32503329 PMCID: PMC7312431 DOI: 10.3390/ijms21114004] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/27/2020] [Accepted: 06/01/2020] [Indexed: 01/10/2023] Open
Abstract
Photodynamic therapy (PDT) has long been known as an effective method for treating surface cancer tissues. Although this technique is widely used in modern medicine, some novel approaches for deep lying tumors have to be developed. Recently, deeper penetration of X-rays into tissues has been implemented, which is now known as X-ray photodynamic therapy (XPDT). The two methods differ in the photon energy used, thus requiring the use of different types of scintillating nanoparticles. These nanoparticles are known to convert the incident energy into the activation energy of a photosensitizer, which leads to the generation of reactive oxygen species. Since not all photosensitizers are found to be suitable for the currently used scintillating nanoparticles, it is necessary to find the most effective biocompatible combination of these two agents. The most successful combinations of nanoparticles for XPDT are presented. Nanomaterials such as metal-organic frameworks having properties of photosensitizers and scintillation nanoparticles are reported to have been used as XPDT agents. The role of metal-organic frameworks for applying XPDT as well as the mechanism underlying the generation of reactive oxygen species are discussed.
Collapse
Affiliation(s)
- Zaira Gadzhimagomedova
- The Smart Materials Research Institute, Southern Federal University, 344090 Rostov-on-Don, Russia; (D.K.); (A.S.)
- Correspondence:
| | - Peter Zolotukhin
- Academy of Biology and Biotechnology, Southern Federal University, 344090 Rostov-on-Don, Russia;
| | - Oleg Kit
- Department of Oncology, National Medical Research Centre for Oncology, 344037 Rostov-on-Don, Russia;
| | - Daria Kirsanova
- The Smart Materials Research Institute, Southern Federal University, 344090 Rostov-on-Don, Russia; (D.K.); (A.S.)
| | - Alexander Soldatov
- The Smart Materials Research Institute, Southern Federal University, 344090 Rostov-on-Don, Russia; (D.K.); (A.S.)
| |
Collapse
|
49
|
Wang M, Yang Q, Li M, Zou H, Wang Z, Ran H, Zheng Y, Jian J, Zhou Y, Luo Y, Ran Y, Jiang S, Zhou X. Multifunctional Nanoparticles for Multimodal Imaging-Guided Low-Intensity Focused Ultrasound/Immunosynergistic Retinoblastoma Therapy. ACS APPLIED MATERIALS & INTERFACES 2020; 12:5642-5657. [PMID: 31940169 DOI: 10.1021/acsami.9b22072] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Retinoblastoma (RB) is prone to delayed diagnosis or treatment and has an increased likelihood of metastasizing. Thus, it is crucial to perform an effective imaging examination and provide optimal treatment of RB to prevent metastasis. Nanoparticles that support diagnostic imaging and targeted therapy are expected to noninvasively integrate tumor diagnosis and treatment. Herein, we report a multifunctional nanoparticle for multimodal imaging-guided low-intensity focused ultrasound (LIFU)/immunosynergistic RB therapy. Magnetic hollow mesoporous gold nanocages (AuNCs) conjugated with Fe3O4 nanoparticles (AuNCs-Fe3O4) were prepared to encapsulate muramyl dipeptide (MDP) and perfluoropentane (PFP). The multimodal imaging capabilities, antitumor effects, and dendritic cell (DC) activation capacity of these nanoparticles combined with LIFU were explored in vitro and in vivo. The biosafety of AuNCs-Fe3O4/MDP/PFP was also evaluated systematically. The multifunctional magnetic nanoparticles enhanced photoacoustic (PA), ultrasound (US), and magnetic resonance (MR) imaging in vivo and in vitro, which was helpful for diagnosis and efficacy evaluation. Upon accumulation in tumors via a magnetic field, the nanoparticles underwent phase transition under LIFU irradiation and MDP was released. A combined effect of AuNCs-Fe3O4/MDP/PFP and LIFU was recorded and verified. AuNCs-Fe3O4/MDP/PFP enhanced the therapeutic effect of LIFU and led to direct apoptosis/necrosis of tumors, while MDP promoted DC maturation and activation and activated the ability of DCs to recognize and clear tumor cells. By enhancing PA/US/MR imaging and inhibiting tumor growth, the multifunctional AuNC-Fe3O4/MDP/PFP nanoparticles show great potential for multimodal imaging-guided LIFU/immunosynergistic therapy of RB. The proposed nanoplatform facilitates cancer theranostics with high biosafety.
Collapse
Affiliation(s)
- Menglei Wang
- Department of Ophthalmology , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Qiming Yang
- Department of Orthopedic , The First Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Meng Li
- Department of Ophthalmology , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Hongmi Zou
- Department of Ophthalmology , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Zhigang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Yuanyi Zheng
- Shanghai Institute of Ultrasound in Medicine , Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai 200233 , P. R. China
| | - Jia Jian
- Department of Ophthalmology , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Yu Zhou
- Department of Ophthalmology , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Yindeng Luo
- Department of Radiology , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Yijun Ran
- Department of Ophthalmology , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Shaoqiu Jiang
- Department of Ophthalmology , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| | - Xiyuan Zhou
- Department of Ophthalmology , The Second Affiliated Hospital of Chongqing Medical University , Chongqing 400010 , P. R. China
| |
Collapse
|