1
|
Ismail M, Wang Y, Li Y, Liu J, Zheng M, Zou Y. Stimuli-Responsive Polymeric Nanocarriers Accelerate On-Demand Drug Release to Combat Glioblastoma. Biomacromolecules 2024; 25:6250-6282. [PMID: 39259212 DOI: 10.1021/acs.biomac.4c00722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Glioblastoma multiforme (GBM) is a highly malignant brain tumor with a poor prognosis and limited treatment options. Drug delivery by stimuli-responsive nanocarriers holds great promise for improving the treatment modalities of GBM. At the beginning of the review, we highlighted the stimuli-active polymeric nanocarriers carrying therapies that potentially boost anti-GBM responses by employing endogenous (pH, redox, hypoxia, enzyme) or exogenous stimuli (light, ultrasonic, magnetic, temperature, radiation) as triggers for controlled drug release mainly via hydrophobic/hydrophilic transition, degradability, ionizability, etc. Modifying these nanocarriers with target ligands further enhanced their capacity to traverse the blood-brain barrier (BBB) and preferentially accumulate in glioma cells. These unique features potentially lead to more effective brain cancer treatment with minimal adverse reactions and superior therapeutic outcomes. Finally, the review summarizes the existing difficulties and future prospects in stimuli-responsive nanocarriers for treating GBM. Overall, this review offers theoretical guidelines for developing intelligent and versatile stimuli-responsive nanocarriers to facilitate precise drug delivery and treatment of GBM in clinical settings.
Collapse
Affiliation(s)
- Muhammad Ismail
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yibin Wang
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yundong Li
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Jiayi Liu
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Meng Zheng
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yan Zou
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| |
Collapse
|
2
|
Manoharan D, Wang LC, Chen YC, Li WP, Yeh CS. Catalytic Nanoparticles in Biomedical Applications: Exploiting Advanced Nanozymes for Therapeutics and Diagnostics. Adv Healthc Mater 2024; 13:e2400746. [PMID: 38683107 DOI: 10.1002/adhm.202400746] [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: 02/26/2024] [Revised: 04/17/2024] [Indexed: 05/01/2024]
Abstract
Catalytic nanoparticles (CNPs) as heterogeneous catalyst reveals superior activity due to their physio-chemical features, such as high surface-to-volume ratio and unique optical, electric, and magnetic properties. The CNPs, based on their physio-chemical nature, can either increase the reactive oxygen species (ROS) level for tumor and antibacterial therapy or eliminate the ROS for cytoprotection, anti-inflammation, and anti-aging. In addition, the catalytic activity of nanozymes can specifically trigger a specific reaction accompanied by the optical feature change, presenting the feasibility of biosensor and bioimaging applications. Undoubtedly, CNPs play a pivotal role in pushing the evolution of technologies in medical and clinical fields, and advanced strategies and nanomaterials rely on the input of chemical experts to develop. Herein, a systematic and comprehensive review of the challenges and recent development of CNPs for biomedical applications is presented from the viewpoint of advanced nanomaterial with unique catalytic activity and additional functions. Furthermore, the biosafety issue of applying biodegradable and non-biodegradable nanozymes and future perspectives are critically discussed to guide a promising direction in developing span-new nanozymes and more intelligent strategies for overcoming the current clinical limitations.
Collapse
Affiliation(s)
- Divinah Manoharan
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
- Interdisciplinary Research Center on Material and Medicinal Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
| | - Liu-Chun Wang
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
| | - Ying-Chi Chen
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
| | - Wei-Peng Li
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
- 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
| | - Chen-Sheng Yeh
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
- Interdisciplinary Research Center on Material and Medicinal Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 701, Taiwan
| |
Collapse
|
3
|
Tang X, Li Y, Zhu T, Lv L, Liu J. Low-dose X-ray stimulated NO-releasing nanocomposites for closed-loop dual-mode cancer therapy. Biomater Sci 2024; 12:4211-4225. [PMID: 38980700 DOI: 10.1039/d4bm00593g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
X-ray-excited photodynamic therapy (X-PDT) employs X-rays as an energy source, overcoming the light penetration limitations of traditional photodynamic therapy (PDT) but is constrained by high-energy radiation and the hypoxic tumor microenvironment. Low-dose X-ray-excited photodynamic therapy and reduction of mitochondrial oxygen consumption can serve as significant breakthroughs in overcoming these barriers. In this study, NaLuF4:Tb/Gd (15%/5%)@NaYF4 (ScNP) nanoparticles adsorbing the photosensitizer MC540 and loaded with α-(nitrate ester) acid (NEAA) were prepared as low X-ray dose triggered nano-scintillators. The final product obtained was NaLuF4:Tb/Gd (15%/5%)@NaYF4@mSiO2@MC540@NEAA (ScNP-MS@MC540@NEAA) nanocomposites, which exhibited intense green luminescence. X-PDT generates cytotoxic reactive oxygen species (ROS) with minimal ionizing radiation damage. Simultaneously, NEAA reacts with glutathione (GSH) to generate nitric oxide (NO) for gaseous treatment of the damaged mitochondrial respiratory chain to reduce oxygen consumption and alleviate hypoxia, enhancing the X-PDT efficacy and realizing a closed-loop treatment. The superoxide ions (˙O2-) can rapidly react with NO produced to form the highly cytotoxic reactive nitrogen species (RNS) peroxynitrite anion (ONOO-), which exhibits higher cytotoxicity compared to ROS. Furthermore, GSH scavenges toxic ROS and maintains the physiological function of tumor cells. It can induce cancer cell overoxidation and nitrosative stress. This work describes a low-dose X-ray-triggered X-PDT system with total radiation of 50 mGy, which involves GSH consumption, self-supplied NO, mitochondrial damage alleviation, and hypoxia relief to generate ROS and RNS, forming a closed-loop anti-hypoxia dual-mode system with synergistically enhanced anti-tumor effects, without significant biological side effects. It provides a promising platform for deep-seated tumor X-PDT with considerable application prospects.
Collapse
Affiliation(s)
- Xiaoli Tang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China.
| | - Yong Li
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China.
| | - Tao Zhu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China.
| | - Longhao Lv
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China.
| | - Jinliang Liu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China.
| |
Collapse
|
4
|
Policei Marques N, Isikawa MM, Muradova Z, Morris T, Berbeco R, Guidelli EJ. Size-Dependent Blue Emission from Europium-Doped Strontium Fluoride Nanoscintillators for X-Ray-Activated Photodynamic Therapy. Adv Healthc Mater 2024; 13:e2400372. [PMID: 38630101 DOI: 10.1002/adhm.202400372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/04/2024] [Indexed: 04/29/2024]
Abstract
Successful implementation of X-ray-activated photodynamic therapy (X-PDT) is challenging because most photosensitizers (PSs) absorb light in the blue region, but few nanoscintillators produce efficient blue scintillation. Here, efficient blue-emitting SrF2:Eu scintillating nanoparticles (ScNPs) are developed. The optimized synthesis conditions result in cubic nanoparticles with ≈32 nm diameter and blue emission at 416 nm. Coating them with the meso-tetra(n-methyl-4-pyridyl) porphyrin (TMPyP) in a core-shell structure (SrF@TMPyP) results in maximum singlet oxygen (1O2) generation upon X-ray irradiation for nanoparticles with 6TMPyP depositions (SrF@6TMPyP). The 1O2 generation is directly proportional to the dose, does not vary in the low-energy X-ray range (48-160 kVp), but is 21% higher when irradiated with low-energy X-rays than irradiations with higher energy gamma rays. In the clonogenic assay, cancer cells treated with SrF@6TMPyP and exposed to X-rays present a significantly reduced survival fraction compared to the controls. The SrF2:Eu ScNPs and their conjugates stand out as tunable nanoplatforms for X-PDT due to the efficient blue emission from the SrF2:Eu cores; the ability to adjust the scintillation emission in terms of color and intensity by controlling the nanoparticle size; the efficient 1O2 production when conjugated to a PS and the efficacy of killing cancer cells.
Collapse
Affiliation(s)
- Natasha Policei Marques
- Departamento de Física-Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, 14040-901, Brazil
| | - Mileni M Isikawa
- Departamento de Física-Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, 14040-901, Brazil
| | - Zeinaf Muradova
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Toby Morris
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
- Department of Physics and Applied Physics, University of Massachusetts Lowell, Lowell, MA, 01854, USA
| | - Ross Berbeco
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Eder J Guidelli
- Departamento de Física-Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, 14040-901, Brazil
| |
Collapse
|
5
|
Zhang Y, Cheng Y, Zhao Z, Jiang S, Zhang Y, Li J, Huang S, Wang W, Xue Y, Li A, Tao Z, Wu Z, Zhang X. Enhanced Chemoradiotherapy for MRSA-Infected Osteomyelitis Using Immunomodulatory Polymer-Reinforced Nanotherapeutics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2304991. [PMID: 38408365 DOI: 10.1002/adma.202304991] [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: 05/26/2023] [Revised: 12/27/2023] [Indexed: 02/28/2024]
Abstract
The eradication of osteomyelitis caused by methicillin-resistant Staphylococcus aureus (MRSA) poses a significant challenge due to its development of biofilm-induced antibiotic resistance and impaired innate immunity, which often leads to frequent surgical failure. Here, the design, synthesis, and performance of X-ray-activated polymer-reinforced nanotherapeutics that modulate the immunological properties of infectious microenvironments to enhance chemoradiotherapy against multidrug-resistant bacterial deep-tissue infections are reported. Upon X-ray radiation, the proposed polymer-reinforced nanotherapeutic generates reactive oxygen species and reactive nitrogen species. To robustly eradicate MRSA biofilms at deep infection sites, these species can specifically bind to MRSA and penetrate biofilms for enhanced chemoradiotherapy treatment. X-ray-activated nanotherapeutics modulate the innate immunity of macrophages to prevent the recurrence of osteomyelitis. The remarkable anti-infection effects of these nanotherapeutics are validated using a rat osteomyelitis model. This study demonstrates the significant potential of a synergistic chemoradiotherapy and immunotherapy method for treating MRSA biofilm-infected osteomyelitis.
Collapse
Affiliation(s)
- Yufei Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of functional polymer materials College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Yijie Cheng
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of functional polymer materials College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Zhe Zhao
- Department of Surgery of Traditional Chinese Medicine, Tianjin Hospital, Tianjin, 300211, China
| | - Shengpeng Jiang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Yuhan Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Jie Li
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of functional polymer materials College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Siyuan Huang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of functional polymer materials College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Wenbo Wang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of functional polymer materials College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Yun Xue
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of functional polymer materials College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Anran Li
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of functional polymer materials College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Zhen Tao
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhongming Wu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Xinge Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, Tianjin Key Laboratory of functional polymer materials College of Chemistry, Nankai University, Tianjin, 300071, China
| |
Collapse
|
6
|
Wu X, Zhou Z, Li K, Liu S. Nanomaterials-Induced Redox Imbalance: Challenged and Opportunities for Nanomaterials in Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308632. [PMID: 38380505 PMCID: PMC11040387 DOI: 10.1002/advs.202308632] [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: 11/11/2023] [Revised: 01/24/2024] [Indexed: 02/22/2024]
Abstract
Cancer cells typically display redox imbalance compared with normal cells due to increased metabolic rate, accumulated mitochondrial dysfunction, elevated cell signaling, and accelerated peroxisomal activities. This redox imbalance may regulate gene expression, alter protein stability, and modulate existing cellular programs, resulting in inefficient treatment modalities. Therapeutic strategies targeting intra- or extracellular redox states of cancer cells at varying state of progression may trigger programmed cell death if exceeded a certain threshold, enabling therapeutic selectivity and overcoming cancer resistance to radiotherapy and chemotherapy. Nanotechnology provides new opportunities for modulating redox state in cancer cells due to their excellent designability and high reactivity. Various nanomaterials are widely researched to enhance highly reactive substances (free radicals) production, disrupt the endogenous antioxidant defense systems, or both. Here, the physiological features of redox imbalance in cancer cells are described and the challenges in modulating redox state in cancer cells are illustrated. Then, nanomaterials that regulate redox imbalance are classified and elaborated upon based on their ability to target redox regulations. Finally, the future perspectives in this field are proposed. It is hoped this review provides guidance for the design of nanomaterials-based approaches involving modulating intra- or extracellular redox states for cancer therapy, especially for cancers resistant to radiotherapy or chemotherapy, etc.
Collapse
Affiliation(s)
- Xumeng Wu
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin150006China
- Zhengzhou Research InstituteHarbin Institute of TechnologyZhengzhou450046China
| | - Ziqi Zhou
- Zhengzhou Research InstituteHarbin Institute of TechnologyZhengzhou450046China
- School of Medicine and HealthHarbin Institute of TechnologyHarbin150006China
| | - Kai Li
- Zhengzhou Research InstituteHarbin Institute of TechnologyZhengzhou450046China
- School of Medicine and HealthHarbin Institute of TechnologyHarbin150006China
| | - Shaoqin Liu
- School of Life Science and TechnologyHarbin Institute of TechnologyHarbin150006China
- Zhengzhou Research InstituteHarbin Institute of TechnologyZhengzhou450046China
- School of Medicine and HealthHarbin Institute of TechnologyHarbin150006China
| |
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
|
Chen L, Zhang S, Duan Y, Song X, Chang M, Feng W, Chen Y. Silicon-containing nanomedicine and biomaterials: materials chemistry, multi-dimensional design, and biomedical application. Chem Soc Rev 2024; 53:1167-1315. [PMID: 38168612 DOI: 10.1039/d1cs01022k] [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: 01/05/2024]
Abstract
The invention of silica-based bioactive glass in the late 1960s has sparked significant interest in exploring a wide range of silicon-containing biomaterials from the macroscale to the nanoscale. Over the past few decades, these biomaterials have been extensively explored for their potential in diverse biomedical applications, considering their remarkable bioactivity, excellent biocompatibility, facile surface functionalization, controllable synthesis, etc. However, to expedite the clinical translation and the unexpected utilization of silicon-composed nanomedicine and biomaterials, it is highly desirable to achieve a thorough comprehension of their characteristics and biological effects from an overall perspective. In this review, we provide a comprehensive discussion on the state-of-the-art progress of silicon-composed biomaterials, including their classification, characteristics, fabrication methods, and versatile biomedical applications. Additionally, we highlight the multi-dimensional design of both pure and hybrid silicon-composed nanomedicine and biomaterials and their intrinsic biological effects and interactions with biological systems. Their extensive biomedical applications span from drug delivery and bioimaging to therapeutic interventions and regenerative medicine, showcasing the significance of their rational design and fabrication to meet specific requirements and optimize their theranostic performance. Additionally, we offer insights into the future prospects and potential challenges regarding silicon-composed nanomedicine and biomaterials. By shedding light on these exciting research advances, we aspire to foster further progress in the biomedical field and drive the development of innovative silicon-composed nanomedicine and biomaterials with transformative applications in biomedicine.
Collapse
Affiliation(s)
- Liang Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Shanshan Zhang
- Department of Ultrasound Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P. R. China
| | - Yanqiu Duan
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China.
| | - Xinran Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China.
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| |
Collapse
|
9
|
Noghreiyan AV, Soleymanifard S, Sazgarnia A. Design of a novel nanoparticle to use X-ray fluorescence of TiO 2 to induce photodynamic effects in the presence of PpIX. Photodiagnosis Photodyn Ther 2024; 45:103890. [PMID: 37981223 DOI: 10.1016/j.pdpdt.2023.103890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 11/07/2023] [Accepted: 11/07/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND Radiotherapy and photodynamic therapy are the methods of cancer treatment. Although one limitation of photodynamic therapy (PDT) is the limited penetration depth of light through tissue, using X-rays does not have this restriction. Self-lighting nanoparticles can convert X-rays into UV/visible. This study focuses on a newly designed nanostructure containing mesoporous silica nanoparticles (MSN), titanium dioxide nanoparticles (TiO2, anatase grade), and protoporphyrin IX (PpIX) as a photosensitizer to overcome the limitations of photodynamic therapy. METHODS After the synthesis and characterization of Ti-MSN/PpIX@PVP nanostructure, two ROSes (OH* and 1O2) were measured when the nanostructures were irradiated with 100 kV and 6 MV photons. The toxicity of Ti-MSN/PpIX@PVP nanostructure in presence and absence of radiation was investigated on DFW and HT-29 cell lines. The in-vitro experiments were analyzed using the MTT assay and colony count assay. Finally, the effect of light exposure in the presence of Ti-MSN/PpIX@PVP nanostructure on the two cell lines was studied. The in-vitro studies were evaluated using the Synergism Index (Syn) and Dose Enhancement Factor (DEF). RESULTS Based on the FESEM (field emission scanning electron Microscopy) images and DLS (dynamic light scattering) measurements, the size of Ti-MSN/PpIX nanostructure was determined as (35.2 nm) and (168.4 nm), respectively. Based on the spectrofluorimetry results, 100 kV photons produced more ROSes than 6 MV photons. The results of MTT assay and colony formation for X-PDT show Syn >1, except for 100 kV photons for HT-29 cell line. The nanostructure also reduced colony formation induced by X-PDT more effectively when irradiated by 100 kV photons on DFW cells. The results obtained from conventional PDT showed that the ED 50 of the HT-29 cell line was 6 times higher than that of the DFW cell line. CONCLUSION Designing and synthesizing Ti-MSN/PpIX@PVP nanostructures offer a promising strategy for reducing the current challenges in PDT and for developing and advancing X-PDT as an innovative cancer treatment technique.
Collapse
Affiliation(s)
- Atefeh Vejdani Noghreiyan
- Department of Medical Physics Radiobiology and Radiation Protection, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Shokouhozaman Soleymanifard
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Physics Research center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ameneh Sazgarnia
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Physics Research center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
10
|
Alvarez N, Sevilla A. Current Advances in Photodynamic Therapy (PDT) and the Future Potential of PDT-Combinatorial Cancer Therapies. Int J Mol Sci 2024; 25:1023. [PMID: 38256096 PMCID: PMC10815790 DOI: 10.3390/ijms25021023] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Photodynamic therapy (PDT) is a two-stage treatment that implies the use of light energy, oxygen, and light-activated compounds (photosensitizers) to elicit cancerous and precancerous cell death after light activation (phototoxicity). The biophysical, bioengineering aspects and its combinations with other strategies are highlighted in this review, both conceptually and as they are currently applied clinically. We further explore the recent advancements of PDT with the use of nanotechnology, including quantum dots as innovative photosensitizers or energy donors as well as the combination of PDT with radiotherapy and immunotherapy as future promising cancer treatments. Finally, we emphasize the potential significance of organoids as physiologically relevant models for PDT.
Collapse
Affiliation(s)
- Niuska Alvarez
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, 08028 Barcelona, Spain;
| | - Ana Sevilla
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, 08028 Barcelona, Spain;
- Institute of Biomedicine, University of Barcelona (IBUB), 08036 Barcelona, Spain
| |
Collapse
|
11
|
Song Q, Zheng Y, Zhong G, Wang S, He C, Li M. Application of Nanoparticles in the Diagnosis and Treatment of Colorectal Cancer. Anticancer Agents Med Chem 2024; 24:1305-1326. [PMID: 39129164 PMCID: PMC11497148 DOI: 10.2174/0118715206323900240807110122] [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: 05/15/2024] [Revised: 06/26/2024] [Accepted: 07/12/2024] [Indexed: 08/13/2024]
Abstract
Colorectal cancer is a common malignant tumor with high morbidity and mortality rates, imposing a huge burden on both patients and the healthcare system. Traditional treatments such as surgery, chemotherapy and radiotherapy have limitations, so finding more effective diagnostic and therapeutic tools is critical to improving the survival and quality of life of colorectal cancer patients. While current tumor targeting research mainly focuses on exploring the function and mechanism of molecular targets and screening for excellent drug targets, it is crucial to test the efficacy and mechanism of tumor cell therapy that targets these molecular targets. Selecting the appropriate drug carrier is a key step in effectively targeting tumor cells. In recent years, nanoparticles have gained significant interest as gene carriers in the field of colorectal cancer diagnosis and treatment due to their low toxicity and high protective properties. Nanoparticles, synthesized from natural or polymeric materials, are NM-sized particles that offer advantages such as low toxicity, slow release, and protection of target genes during delivery. By modifying nanoparticles, they can be targeted towards specific cells for efficient and safe targeting of tumor cells. Numerous studies have demonstrated the safety, efficiency, and specificity of nanoparticles in targeting tumor cells, making them a promising gene carrier for experimental and clinical studies. This paper aims to review the current application of nanoparticles in colorectal cancer diagnosis and treatment to provide insights for targeted therapy for colorectal cancer while also highlighting future prospects for nanoparticle development.
Collapse
Affiliation(s)
- Qiuyu Song
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yifeng Zheng
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guoqiang Zhong
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shanping Wang
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chengcheng He
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mingsong Li
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
12
|
Gadzhimagomedova Z, Pankin I, Polyakov V, Khodakova D, Medvedev P, Zelenikhin P, Shamsutdinov N, Chapek S, Goncharova A, Soldatov A. Single-Stage Microfluidic Synthesis Route for BaGdF 5:Tb 3+-Based Nanocomposite Materials: Synthesis, Characterization and Biodistribution. Int J Mol Sci 2023; 24:17159. [PMID: 38138988 PMCID: PMC10742823 DOI: 10.3390/ijms242417159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
Rare-earth-doped nanoscaled BaGdF5 is known as an efficient contrasting agent for X-ray micro-CT and NMR as well as a promising candidate for X-ray photodynamic therapy, thereby opening an opportunity for theragnostic applications. Conventional synthesis of Ln-doped BaGdF5 consider a long-lasting batch procedure, while a conjugation with photosensitizer usually implies a separate stage requiring active mixing. To the best of our knowledge, in this work, we for the first time obtain BaGdF5:Tb3+ nanophosphors in a microfluidic route at temperatures as low as 100 °C while decreasing the time of thermal treatment down to 6 min. The proposed synthesis route allows for the obtaining of single-phase and monodisperse BaGd1-xF5:Tbx3+ nanoparticles with an averaged particle size of ca. 7-9 nm and hydrodynamic radius around 22 nm, as estimated from TEM and DLS, respectively. In addition, X-ray-excited optical luminescence has been recorded in situ for the series of nanophosphors synthesis with varied flow rates of Tb3+ and Gd3+ stock solutions, thereby anticipating a possible application of microfluidics for screening a wide range of possible co-dopants and reaction conditions and its effect on the optical properties of the synthesized materials. Moreover, we demonstrated that BaGd1-xF5:Tbx3+@RoseBengal conjugates might be obtained in a single-stage route by implementing an additional mixer at the synthesis outcome, namely, by mixing the resulting reaction mixture containing nanoparticles with an equivalent flow of photosensitizer aqueous solution. In vitro cytotoxicity test declares moderate toxicity effect on different cell lines, while the results of flow cytometry indirectly confirm cellular uptake. Finally, we report long-term biodistribution monitoring of the synthesized nanocomposites assessed by X-ray micro-CT in the in vivo experiments on balb/c mice, which depicts an unusual character of agents' accumulation.
Collapse
Affiliation(s)
- Zaira Gadzhimagomedova
- The Smart Materials Research Institute, Southern Federal University, 344090 Rostov-on-Don, Russia; (Z.G.); (P.M.); (S.C.); (A.S.)
| | - Ilia Pankin
- The Smart Materials Research Institute, Southern Federal University, 344090 Rostov-on-Don, Russia; (Z.G.); (P.M.); (S.C.); (A.S.)
| | - Vladimir Polyakov
- The Smart Materials Research Institute, Southern Federal University, 344090 Rostov-on-Don, Russia; (Z.G.); (P.M.); (S.C.); (A.S.)
| | - Darya Khodakova
- National Medical Research Centre for Oncology, 344037 Rostov-on-Don, Russia; (D.K.); (A.G.)
| | - Pavel Medvedev
- The Smart Materials Research Institute, Southern Federal University, 344090 Rostov-on-Don, Russia; (Z.G.); (P.M.); (S.C.); (A.S.)
| | - Pavel Zelenikhin
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (P.Z.); (N.S.)
| | - Nail Shamsutdinov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia; (P.Z.); (N.S.)
| | - Sergey Chapek
- The Smart Materials Research Institute, Southern Federal University, 344090 Rostov-on-Don, Russia; (Z.G.); (P.M.); (S.C.); (A.S.)
| | - Anna Goncharova
- National Medical Research Centre for Oncology, 344037 Rostov-on-Don, Russia; (D.K.); (A.G.)
| | - Alexander Soldatov
- The Smart Materials Research Institute, Southern Federal University, 344090 Rostov-on-Don, Russia; (Z.G.); (P.M.); (S.C.); (A.S.)
| |
Collapse
|
13
|
Zhang D, Liu D, Wang C, Su Y, Zhang X. Nanoreactor-based catalytic systems for therapeutic applications: Principles, strategies, and challenges. Adv Colloid Interface Sci 2023; 322:103037. [PMID: 37931381 DOI: 10.1016/j.cis.2023.103037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/25/2023] [Accepted: 10/29/2023] [Indexed: 11/08/2023]
Abstract
Inspired by natural catalytic compartments, various synthetic compartments that seclude catalytic reactions have been developed to understand complex multistep biosynthetic pathways, bestow therapeutic effects, or extend biosynthetic pathways in living cells. These emerging nanoreactors possessed many advantages over conventional biomedicine, such as good catalytic activity, specificity, and sustainability. In the past decade, a great number of efficient catalytic systems based on diverse nanoreactors (polymer vesicles, liposome, polymer micelles, inorganic-organic hybrid materials, MOFs, etc.) have been designed and employed to initiate in situ catalyzed chemical reactions for therapy. This review aims to present the recent progress in the development of catalytic systems based on nanoreactors for therapeutic applications, with a special emphasis on the principles and design strategies. Besides, the key components of nanoreactor-based catalytic systems, including nanocarriers, triggers or energy inputs, and products, are respectively introduced and discussed in detail. Challenges and prospects in the fabrication of therapeutic catalytic nanoreactors are also discussed as a conclusion to this review. We believe that catalytic nanoreactors will play an increasingly important role in modern biomedicine, with improved therapeutic performance and minimal side effects.
Collapse
Affiliation(s)
- Dan Zhang
- Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| | - Dongcheng Liu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
| | - Chunfei Wang
- Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| | - Yanhong Su
- Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| | - Xuanjun Zhang
- Faculty of Health Sciences, University of Macau, Macau SAR 999078, China; MOE Frontiers Science Centre for Precision Oncology, University of Macau, Macau SAR 999078, China.
| |
Collapse
|
14
|
Babu B, Stoltz SA, Mittal A, Pawar S, Kolanthai E, Coathup M, Seal S. Inorganic Nanoparticles as Radiosensitizers for Cancer Treatment. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:2873. [PMID: 37947718 PMCID: PMC10647410 DOI: 10.3390/nano13212873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023]
Abstract
Nanotechnology has expanded what can be achieved in our approach to cancer treatment. The ability to produce and engineer functional nanoparticle formulations to elicit higher incidences of tumor cell radiolysis has resulted in substantial improvements in cancer cell eradication while also permitting multi-modal biomedical functionalities. These radiosensitive nanomaterials utilize material characteristics, such as radio-blocking/absorbing high-Z atomic number elements, to mediate localized effects from therapeutic irradiation. These materials thereby allow subsequent scattered or emitted radiation to produce direct (e.g., damage to genetic materials) or indirect (e.g., protein oxidation, reactive oxygen species formation) damage to tumor cells. Using nanomaterials that activate under certain physiologic conditions, such as the tumor microenvironment, can selectively target tumor cells. These characteristics, combined with biological interactions that can target the tumor environment, allow for localized radio-sensitization while mitigating damage to healthy cells. This review explores the various nanomaterial formulations utilized in cancer radiosensitivity research. Emphasis on inorganic nanomaterials showcases the specific material characteristics that enable higher incidences of radiation while ensuring localized cancer targeting based on tumor microenvironment activation. The aim of this review is to guide future research in cancer radiosensitization using nanomaterial formulations and to detail common approaches to its treatment, as well as their relations to commonly implemented radiotherapy techniques.
Collapse
Affiliation(s)
- Balaashwin Babu
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
| | - Samantha Archer Stoltz
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Agastya Mittal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Shreya Pawar
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Elayaraja Kolanthai
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
| | - Melanie Coathup
- Biionix Cluster, University of Central Florida, Orlando, FL 32827, USA;
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Sudipta Seal
- Advanced Materials Processing and Analysis Center, Department of Materials Science and Engineering, University of Central Florida, Orlando, FL 32826, USA; (B.B.); (S.A.S.); (A.M.); (S.P.); (E.K.)
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- Nanoscience Technology Center, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
15
|
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: 1.0] [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
|
16
|
Ding S, Chen L, Liao J, Huo Q, Wang Q, Tian G, Yin W. Harnessing Hafnium-Based Nanomaterials for Cancer Diagnosis and Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2300341. [PMID: 37029564 DOI: 10.1002/smll.202300341] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/01/2023] [Indexed: 06/19/2023]
Abstract
With the rapid development of nanotechnology and nanomedicine, there are great interests in employing nanomaterials to improve the efficiency of disease diagnosis and treatment. The clinical translation of hafnium oxide (HfO2 ), commercially namedas NBTXR3, as a new kind of nanoradiosensitizer for radiotherapy (RT) of cancers has aroused extensive interest in researches on Hf-based nanomaterials for biomedical application. In the past 20 years, Hf-based nanomaterials have emerged as potential and important nanomedicine for computed tomography (CT)-involved bioimaging and RT-associated cancer treatment due to their excellent electronic structures and intrinsic physiochemical properties. In this review, a bibliometric analysis method is employed to summarize the progress on the synthesis technology of various Hf-based nanomaterials, including HfO2 , HfO2 -based compounds, and Hf-organic ligand coordination hybrids, such as metal-organic frameworks or nanoscaled coordination polymers. Moreover, current states in the application of Hf-based CT-involved contrasts for tissue imaging or cancer diagnosis are reviewed in detail. Importantly, the recent advances in Hf-based nanomaterials-mediated radiosensitization and synergistic RT with other current mainstream treatments are also generalized. Finally, current challenges and future perspectives of Hf-based nanomaterials with a view to maximize their great potential in the research of translational medicine are also discussed.
Collapse
Affiliation(s)
- Shuaishuai Ding
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of Tumor Immunopathology, Ministry of Education of China, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
| | - Lei Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jing Liao
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of Tumor Immunopathology, Ministry of Education of China, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
- Laboratory for Micro-sized Functional Materials, Department of Chemistry and College of Elementary Education, Capital Normal University, Beijing, 100048, P. R. China
| | - Qing Huo
- College of Biochemical and Engineering, Beijing Union University, Beijing, 100023, China
| | - Qiang Wang
- Laboratory for Micro-sized Functional Materials, Department of Chemistry and College of Elementary Education, Capital Normal University, Beijing, 100048, P. R. China
| | - Gan Tian
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of Tumor Immunopathology, Ministry of Education of China, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P. R. China
- Chongqing Institute of Advanced Pathology, Jinfeng Laboratory, Chongqing, 401329, P. R. China
| | - Wenyan Yin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
17
|
Dev Tripathi A, Katiyar S, Mishra A. Glypican1: a potential cancer biomarker for nanotargeted therapy. Drug Discov Today 2023:103660. [PMID: 37301249 DOI: 10.1016/j.drudis.2023.103660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/11/2023] [Accepted: 06/05/2023] [Indexed: 06/12/2023]
Abstract
Glypicans (GPCs) are generally involved in cellular signaling, growth and proliferation. Previous studies reported their roles in cancer proliferation. GPC1 is a co-receptor for a variety of growth-related ligands, thereby stimulating the tumor microenvironment by promoting angiogenesis and epithelial-mesenchymal transition (EMT). This work reviews GPC1-biomarker-assisted drug discovery by the application of nanostructured materials, creating nanotheragnostics for targeted delivery and application in liquid biopsies. The review includes details of GPC1 as a potential biomarker in cancer progression as well as a potential candidate for nano-mediated drug discovery.
Collapse
Affiliation(s)
- Abhay Dev Tripathi
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi-221005, India
| | - Soumya Katiyar
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi-221005, India
| | - Abha Mishra
- School of Biochemical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi-221005, India.
| |
Collapse
|
18
|
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: 5.0] [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
|
19
|
Zhen W, Weichselbaum RR, Lin W. Nanoparticle-Mediated Radiotherapy Remodels the Tumor Microenvironment to Enhance Antitumor Efficacy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2206370. [PMID: 36524978 PMCID: PMC10213153 DOI: 10.1002/adma.202206370] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 12/12/2022] [Indexed: 05/26/2023]
Abstract
Radiotherapy (RT) uses ionizing radiation to eradicate localized tumors and, in rare cases, control tumors outside of the irradiated fields via stimulating an antitumor immune response (abscopal effect). However, the therapeutic effect of RT is often limited by inherent physiological barriers of the tumor microenvironment (TME), such as hypoxia, abnormal vasculature, dense extracellular matrix (ECM), and an immunosuppressive TME. Thus, it is critical to develop new RT strategies that can remodel the TME to overcome radio-resistance and immune suppression. In the past decade, high-Z-element nanoparticles have been developed to increase radiotherapeutic indices of localized tumors by reducing X-ray doses and side effects to normal tissues and enhance abscopal effects by activating the TME to elicit systemic antitumor immunity. In this review, the principles of RT and radiosensitization, the mechanisms of radio-resistance and immune suppression, and the use of various nanoparticles to sensitize RT and remodel TMEs for enhanced antitumor efficacy are discussed. The challenges in clinical translation of multifunctional TME-remodeling nanoradiosensitizers are also highlighted.
Collapse
Affiliation(s)
- Wenyao Zhen
- Department of Chemistry, Department of Radiation and Cellular Oncology, and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| | - Wenbin Lin
- Department of Chemistry, Department of Radiation and Cellular Oncology, and the Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
20
|
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: 2.0] [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
|
21
|
Shu G, Zhao H, Zhang X. Persistent luminescent metal-organic framework nanocomposite enables autofluorescence-free dual modal imaging-guided drug delivery. Biomater Sci 2023; 11:1797-1809. [PMID: 36655655 DOI: 10.1039/d2bm01920e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Molecular imaging-guided therapy was essential for realizing precise cancer intervention, while designing an imaging platform to achieve autofluorescence-free imaging for dual modal imaging-guided drug delivery remains a challenge. Near-infrared persistent luminescence nanoparticles (NIR PLNPs) were promising for tumor imaging due to no background interference from the tissue. Herein, a persistent luminescent metal-organic framework (PLNPs@MIL-100(Fe)) is prepared via a layer-by-layer method for dual-modal imaging-guided drug delivery. The PLNPs@MIL-100(Fe) exhibit NIR persistent luminescence emitting and T2-weighted signal, achieving precise in vivo dual-modal imaging of tumor-bearing mice by providing high spatial resolution MR imaging and autofluorescence-free NIR imaging. The porous MIL-100(Fe) shell provides PLNPs@MIL-100(Fe) with up to 87.1% drug loading capacity and acid-triggered drug release for drug delivery. We envision that the proposed PLNPs@MIL-100(Fe) platform would provide an effective approach for precise tumor imaging and versatile drug delivery.
Collapse
Affiliation(s)
- Gang Shu
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China.
| | - Huaixin Zhao
- Frontier Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (MOE), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, P.R. China.
| | - Xuening Zhang
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China.
| |
Collapse
|
22
|
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: 5.0] [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
|
23
|
Souris JS, Leoni L, Zhang HJ, Pan A, Tanios E, Tsai HM, Balyasnikova IV, Bissonnette M, Chen CT. X-ray Activated Nanoplatforms for Deep Tissue Photodynamic Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:673. [PMID: 36839041 PMCID: PMC9962876 DOI: 10.3390/nano13040673] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/12/2023] [Accepted: 02/01/2023] [Indexed: 05/10/2023]
Abstract
Photodynamic therapy (PDT), the use of light to excite photosensitive molecules whose electronic relaxation drives the production of highly cytotoxic reactive oxygen species (ROS), has proven an effective means of oncotherapy. However, its application has been severely constrained to superficial tissues and those readily accessed either endoscopically or laparoscopically, due to the intrinsic scattering and absorption of photons by intervening tissues. Recent advances in the design of nanoparticle-based X-ray scintillators and photosensitizers have enabled hybridization of these moieties into single nanocomposite particles. These nanoplatforms, when irradiated with diagnostic doses and energies of X-rays, produce large quantities of ROS and permit, for the first time, non-invasive deep tissue PDT of tumors with few of the therapeutic limitations or side effects of conventional PDT. In this review we examine the underlying principles and evolution of PDT: from its initial and still dominant use of light-activated, small molecule photosensitizers that passively accumulate in tumors, to its latest development of X-ray-activated, scintillator-photosensitizer hybrid nanoplatforms that actively target cancer biomarkers. Challenges and potential remedies for the clinical translation of these hybrid nanoplatforms and X-ray PDT are also presented.
Collapse
Affiliation(s)
- Jeffrey S. Souris
- Department of Radiology, The University of Chicago, Chicago, IL 60637, USA
- Integrated Small Animal Imaging Research Resource, Office of Shared Research Facilities, The University of Chicago, Chicago, IL 60637, USA
| | - Lara Leoni
- Integrated Small Animal Imaging Research Resource, Office of Shared Research Facilities, The University of Chicago, Chicago, IL 60637, USA
| | - Hannah J. Zhang
- Department of Radiology, The University of Chicago, Chicago, IL 60637, USA
- Integrated Small Animal Imaging Research Resource, Office of Shared Research Facilities, The University of Chicago, Chicago, IL 60637, USA
| | - Ariel Pan
- Department of Radiology, The University of Chicago, Chicago, IL 60637, USA
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY 10065, USA
| | - Eve Tanios
- Department of Radiology, The University of Chicago, Chicago, IL 60637, USA
| | - Hsiu-Ming Tsai
- Integrated Small Animal Imaging Research Resource, Office of Shared Research Facilities, The University of Chicago, Chicago, IL 60637, USA
| | | | - Marc Bissonnette
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Chin-Tu Chen
- Department of Radiology, The University of Chicago, Chicago, IL 60637, USA
- Integrated Small Animal Imaging Research Resource, Office of Shared Research Facilities, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
24
|
Zhang L, Xu F, Lei T, Zhang X, Lan B, Li T, Yu J, Lu H, Zhang W. Growth Phase Diagram and X-ray Excited Luminescence Properties of NaLuF4:Tb3+ Nanoparticles. ARAB J CHEM 2023. [DOI: 10.1016/j.arabjc.2023.104672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
|
25
|
Hong Z, Chen Z, Chen Q, Yang H. Advancing X-ray Luminescence for Imaging, Biosensing, and Theragnostics. Acc Chem Res 2023; 56:37-51. [PMID: 36533853 DOI: 10.1021/acs.accounts.2c00517] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
X-ray luminescence is an optical phenomenon in which chemical compounds known as scintillators can emit short-wavelength light upon the excitation of X-ray photons. Since X-rays exhibit well-recognized advantages of deep penetration toward tissues and a minimal autofluorescence background in biological samples, X-ray luminescence has been increasingly becoming a promising optical tool for tackling the challenges in the fields of imaging, biosensing, and theragnostics. In recent years, the emergence of nanocrystal scintillators have further expanded the application scenarios of X-ray luminescence, such as high-resolution X-ray imaging, autofluorescence-free detection of biomarkers, and noninvasive phototherapy in deep tissues. Meanwhile, X-ray luminescence holds great promise in breaking the depth dependency of deep-seated lesion treatment and achieving synergistic radiotherapy with phototherapy.In this Account, we provide an overview of recent advances in developing advanced X-ray luminescence for applications in imaging, biosensing, theragnostics, and optogenetics neuromodulation. We first introduce solution-processed lead halide all-inorganic perovskite nanocrystal scintillators that are able to convert X-ray photons to multicolor X-ray luminescence. We have developed a perovskite nanoscintillator-based X-ray detector for high-resolution X-ray imaging of the internal structure of electronic circuits and biological samples. We further advanced the development of flexible X-ray luminescence imaging using solution-processable lanthanide-doped nanoscintillators featuring long-lived X-ray luminescence to image three-dimensional irregularly shaped objects. We also outline the general principles of high-contrast in vivo X-ray luminescence imaging which combines nanoscintillators with functional biomolecules such as aptamers, peptides, and antibodies. High-quality X-ray luminescence nanoprobes were engineered to achieve the high-sensitivity detection of various biomarkers, which enabled the avoidance of interference from the biological matrix autofluorescence and photon scattering. By marrying X-ray luminescence probes with stimuli-responsive materials, multifunctional theragnostic nanosystems were constructed for on-demand synergistic gas radiotherapy with excellent therapeutic effects. By taking advantage of the capability of X-rays to penetrate the skull, we also demonstrated the development of controllable, wireless optogenetic neuromodulation using X-ray luminescence probes while obviating damage from traditional optical fibers. Furthermore, we discussed in detail some challenges and future development of X-ray luminescence in terms of scintillator synthesis and surface modification, mechanism studies, and their other potential applications to provide useful guidance for further advancing the development of X-ray luminescence.
Collapse
Affiliation(s)
- Zhongzhu Hong
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, P. R. China
| | - Zhaowei Chen
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, P. R. China
| | - Qiushui Chen
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, P. R. China.,Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou 350108, P. R. China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou 350108, P. R. China.,Fujian Science & Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou 350108, P. R. China
| |
Collapse
|
26
|
Sun W, Chu C, Li S, Ma X, Liu P, Chen S, Chen H. Nanosensitizer-mediated unique dynamic therapy tactics for effective inhibition of deep tumors. Adv Drug Deliv Rev 2023; 192:114643. [PMID: 36493905 DOI: 10.1016/j.addr.2022.114643] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/08/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
X-ray and ultrasound waves are widely employed for diagnostic and therapeutic purposes in clinic. Recently, they have been demonstrated to be ideal excitation sources that activate sensitizers for the dynamic therapy of deep-seated tumors due to their excellent tissue penetration. Here, we focused on the recent progress in five years in the unique dynamic therapy strategies for the effective inhibition of deep tumors that activated by X-ray and ultrasound waves. The concepts, mechanisms, and typical nanosensitizers used as energy transducers are described as well as their applications in oncology. The future developments and potential challenges are also discussed. These unique therapeutic methods are expected to be developed as depth-independent, minimally invasive, and multifunctional strategies for the clinic treatment of various deep malignancies.
Collapse
Affiliation(s)
- Wenjing Sun
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 311200, China; State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Chengchao Chu
- Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Engineering Research Center of Eye Regenerative Medicine, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Shi Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xiaoqian Ma
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Peifei Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Shileng Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hongmin Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
27
|
Xian T, Meng Q, Gao F, Hu M, Wang X. Functionalization of luminescent lanthanide complexes for biomedical applications. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
28
|
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: 5.5] [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
|
29
|
Enhanced Photodynamic Therapy: A Review of Combined Energy Sources. Cells 2022; 11:cells11243995. [PMID: 36552759 PMCID: PMC9776440 DOI: 10.3390/cells11243995] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/04/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Photodynamic therapy (PDT) has been used in recent years as a non-invasive treatment for cancer, due to the side effects of traditional treatments such as surgery, radiotherapy, and chemotherapy. This therapeutic technique requires a photosensitizer, light energy, and oxygen to produce reactive oxygen species (ROS) which mediate cellular toxicity. PDT is a useful non-invasive therapy for cancer treatment, but it has some limitations that need to be overcome, such as low-light-penetration depths, non-targeting photosensitizers, and tumor hypoxia. This review focuses on the latest innovative strategies based on the synergistic use of other energy sources, such as non-visible radiation of the electromagnetic spectrum (microwaves, infrared, and X-rays), ultrasound, and electric/magnetic fields, to overcome PDT limitations and enhance the therapeutic effect of PDT. The main principles, mechanisms, and crucial elements of PDT are also addressed.
Collapse
|
30
|
Hamdy NM, Eskander G, Basalious EB. Insights on the Dynamic Innovative Tumor Targeted-Nanoparticles-Based Drug Delivery Systems Activation Techniques. Int J Nanomedicine 2022; 17:6131-6155. [PMID: 36514378 PMCID: PMC9741821 DOI: 10.2147/ijn.s386037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
Anti-cancer conventional chemotherapeutic drugs novel formula progress, nowadays, uses nano technology for targeted drug delivery, specifically tailored to overcome therapeutic agents' delivery challenges. Polymer drug delivery systems (DDS) play a crucial role in minimizing off-target side effects arising when using standard cytotoxic drugs. Using nano-formula for targeted localized action, permits using larger effective cytotoxic doses on a single special spot, that can seriously cause harm if it was administered systemically. Therefore, various nanoparticles (NPs) specifically have attached groups for targeting capabilities, not seen in bulk materials, which then need activation. In this review, we will present a simple innovative, illustrative, in a cartoon-way, enumeration of NP anti-cancer drug targeting delivery system activation-types. Area(s) covered in this review are the mechanisms of various NP activation techniques.
Collapse
Affiliation(s)
- Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Georgette Eskander
- Faculty of Pharmacy, Ain Shams University, Postgraduate Student, Cairo, Egypt
| | - Emad B Basalious
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
31
|
Polyakov V, Gadzhimagomedova Z, Kirsanova D, Soldatov A. Synthesis Optimization of BaGdF 5:x%Tb 3+ Nanophosphors for Tunable Particle Size. MATERIALS (BASEL, SWITZERLAND) 2022; 15:8559. [PMID: 36500057 PMCID: PMC9740830 DOI: 10.3390/ma15238559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 06/17/2023]
Abstract
X-ray photodynamic therapy (XPDT) is aimed at the treatment of deep-located malignant tumors thanks to the high penetration depth of X-rays. In XPDT therapy, it is necessary to use materials that effectively absorb X-rays and convert them into visible radiation-nanophosphors. Rare-earth elements, fluorides, in particular, doped BaGdF5, are known to serve as efficient nanophosphor. On the other hand, the particle size of nanophosphors has a crucial impact on biodistribution, cell uptake, and cytotoxicity. In this work, we investigated various Tb:Gd ratios in the range from 0.1 to 0.5 and optimized the terbium content to achieve the maximum luminescence under X-ray excitation. The effect of temperature, composition of the ethylene glycol/water solvent, and the synthesis technique (solvothermal and microwave) on the size of the nanophosphors was explored. It was found that the synthesis techniques and the solvent composition had the greatest influence on the averaged particle size. By varying these two parameters, it is possible to tune the size of the nanophosphor particles, which make them suitable for biomedical applications.
Collapse
|
32
|
Jiang H, Wang R, Zhang Q, Song L, Sun X, Shi J, Zhang Y. A dual-functional nanoplatform based on NIR and green dual-emissive persistent luminescence nanoparticles for X-ray excited persistent luminescence imaging and photodynamic therapy. NANOSCALE 2022; 14:15451-15461. [PMID: 36226462 DOI: 10.1039/d2nr03631b] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Persistent luminescence nanoparticles (PLNPs) possess advantages for high-sensitivity bioimaging and continuous photodynamic therapy (PDT) because they can emit persistent luminescence (PerL) after excitation ceases. However, PLNPs are limited to single-wavelength emission, which can only efficiently realize one of the functions of bioimaging or PDT. In addition, most PLNPs are excited by shallow tissue penetrating excitation light, which makes it difficult to achieve repeatable in vivo applications with high efficiency. Herein, X-ray-excited PLNPs (Zn3Ga2Ge2O10:Cr3+,Mn2+, ZGGCM) with dual emission for in vivo X-rays repeatedly activated PerL imaging and tumor PDT are reported for the first time. ZGGCM exhibits dual-emission peaks after X-ray excitation/re-excitation, located at 698 nm and 532 nm, respectively. Additionally, ZGGCM is modified with the photosensitizer rose bengal (RB) to construct a dual-functional nanoplatform based on PerL imaging and PDT. The results indicate that the PerL emission peak (698 nm) of Cr3+ ions in ZGGCM possesses excellent near-infrared (NIR) PerL imaging performance, and the green PerL emission peak (532 nm) of Mn2+ ions can activate RB effectively and generate reactive oxygen species (ROS), thereby causing a significant antitumor effect. This unique dual-functional nanoplatform is expected to further promote the application of PLNPs in the integration of efficient tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Huimin Jiang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China.
- Ganjiang Innovation Academy, Chinese Academy of Sciences, Ganzhou, 341000, China
| | - Ruoping Wang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qian Zhang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China.
- Ganjiang Innovation Academy, Chinese Academy of Sciences, Ganzhou, 341000, China
| | - Liang Song
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China.
- Ganjiang Innovation Academy, Chinese Academy of Sciences, Ganzhou, 341000, China
| | - Xia Sun
- Fujian Science and Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou 350108, P.R. China
| | - Junpeng Shi
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China.
- Ganjiang Innovation Academy, Chinese Academy of Sciences, Ganzhou, 341000, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yun Zhang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China.
- Ganjiang Innovation Academy, Chinese Academy of Sciences, Ganzhou, 341000, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
33
|
|
34
|
Chen YC, Liu YJ, Lee CL, Pham KY, Manoharan D, Thangudu S, Su CH, Yeh CS. Engineering H 2 O 2 and O 2 Self-Supplying Nanoreactor to Conduct Synergistic Chemiexcited Photodynamic and Calcium-Overloaded Therapy in Orthotopic Hepatic Tumors. Adv Healthc Mater 2022; 11:e2201613. [PMID: 35879269 DOI: 10.1002/adhm.202201613] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Indexed: 01/28/2023]
Abstract
Photodynamic therapy (PDT) is traditionally ineffective for deeply embedded tumors due to the poor penetration depth of the excitation light. Chemiluminescence resonance energy transfer (CRET) has emerged as a promising mode of PDT without external light. To date, related research has frequently used endogenous hydrogen peroxide (H2 O2 ) and oxygen (O2 ) inside the solid tumor microenvironment to trigger CRET-mediated PDT. Unfortunately, this significantly restricts treatment efficacy and the development of further biomedical applications because of the limited amounts of endogenous H2 O2 and O2 . Herein, a nanohybrid (mSiO2 /CaO2 /CPPO/Ce6: mSCCC) nanoparticle (NP) is designed to achieve synergistic CRET-mediated PDT and calcium (Ca2+ )-overload-mediated therapy. The calcium peroxide (CaO2 ) formed inside mesoporous SiO2 (mSC) with the inclusion of the chemiluminescent agent (CPPO) and photosensitizer (Ce6) self-supplies H2 O2 , O2 , and Ca2+ allowing for the subsequent treatments. The Ce6 in mSCCC NPs is excited by chemical energy in situ following the supply of H2 O2 and O2 to produce singlet oxygen (1 O2 ). The nanohybrid NPs are coated with stearic acid to avoid decomposition during blood circulation through contact with aqueous environment. This nanohybrid shows promising performance in the generation of 1 O2 for external light-free PDT and the release of Ca2+ ions for Ca2+ -overloaded therapy against orthotopic hepatocellular carcinoma.
Collapse
Affiliation(s)
- Ying-Chi Chen
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
| | - Yu-Ju Liu
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
| | - Chin-Lai Lee
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 833, Taiwan
| | - Khang-Yen Pham
- Department of Chemistry, University of Education, Hue University, Hue City, 530000, Vietnam
| | - Divinah Manoharan
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
| | - Suresh Thangudu
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 833, Taiwan
| | - Chia-Hao Su
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, 833, Taiwan.,Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan.,Center for General Education, Chang Gung University, Taoyuan, 333, Taiwan
| | - Chen-Sheng Yeh
- Department of Chemistry, National Cheng Kung University, Tainan, 701, Taiwan
| |
Collapse
|
35
|
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
|
36
|
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
|
37
|
Jiang F, Lee C, Zhang W, Jiang W, Cao Z, Chong HB, Yang W, Zhan S, Li J, Teng Y, Li Z, Xie J. Radiodynamic therapy with CsI(na)@MgO nanoparticles and 5-aminolevulinic acid. J Nanobiotechnology 2022; 20:330. [PMID: 35842630 PMCID: PMC9288050 DOI: 10.1186/s12951-022-01537-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/16/2022] [Indexed: 12/06/2022] Open
Abstract
Background Radiodynamic therapy (RDT) holds the potential to overcome the shallow tissue penetration issue associated with conventional photodynamic therapy (PDT). To this end, complex and sometimes toxic scintillator–photosensitizer nanoconjugates are often used, posing barriers for large-scale manufacturing and regulatory approval. Methods Herein, we report a streamlined RDT strategy based on CsI(Na)@MgO nanoparticles and 5-aminolevulinic acid (5-ALA). 5-ALA is a clinically approved photosensitizer, converted to protoporphyrin IX (PpIX) in cancer cells’ mitochondria. CsI(Na)@MgO nanoparticles produce strong ~ 410 nm X-ray luminescence, which matches the Soret band of PpIX. We hypothesize that the CsI(Na)@MgO-and-5-ALA combination can mediate RDT wherein mitochondria-targeted PDT synergizes with DNA-targeted irradiation for efficient cancer cell killing. Because scintillator nanoparticles and photosensitizer are administered separately, the approach forgoes issues such as self-quenching or uncontrolled release of photosensitizers. Results When tested in vitro with 4T1 cells, the CsI(Na)@MgO and 5-ALA combination elevated radiation-induced reactive oxygen species (ROS), enhancing damages to mitochondria, DNA, and lipids, eventually reducing cell proliferation and clonogenicity. When tested in vivo in 4T1 models, RDT with the CsI(Na)@MgO and 5-ALA combination significantly improved tumor suppression and animal survival relative to radiation therapy (RT) alone. After treatment, the scintillator nanoparticles, made of low-toxic alkali and halide elements, were efficiently excreted, causing no detectable harm to the hosts. Conclusions Our studies show that separately administering CsI(Na)@MgO nanoparticles and 5-ALA represents a safe and streamlined RDT approach with potential in clinical translation. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01537-z.
Collapse
Affiliation(s)
- Fangchao Jiang
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA
| | - Chaebin Lee
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA
| | - Weizhong Zhang
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA
| | - Wen Jiang
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA
| | - Zhengwei Cao
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA
| | | | - Wei Yang
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA
| | - Shuyue Zhan
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA
| | - Jianwen Li
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA
| | - Yong Teng
- Department of Hematology and Medical Oncology & Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Zibo Li
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Jin Xie
- Department of Chemistry, University of Georgia, Athens, GA, 30602, USA.
| |
Collapse
|
38
|
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: 8.5] [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
|
39
|
Chen L, Zhang J, Xu L, Zhu L, Jing J, Feng Y, Wang Z, Liu P, Sun W, Liu X, Li Y, Chen H. Composition tunability of semiconductor radiosensitizers for low-dose X-ray induced photodynamic therapy. J Nanobiotechnology 2022; 20:293. [PMID: 35729553 PMCID: PMC9210653 DOI: 10.1186/s12951-022-01494-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/01/2022] [Indexed: 11/29/2022] Open
Abstract
Radiation therapy is one of the most commonly used methods in clinical cancer treatment, and radiosensitizers could achieve enhanced therapeutic efficacy by incorporating heavy elements into structures. However, the secondary excitation of these high-Z elements-doped nanosensitizers still imply intrinsic defects of low efficiency. Herein, we designed Bi-doped titanium dioxide nanosensitizers in which high-Z Bi ions with adjustable valence state (Bi3+ or Bi4+) replaced some positions of Ti4+ of anatase TiO2, increasing both X-rays absorption and oxygen vacancies. The as-prepared TiO2:Bi nanosensitizers indicated high ionizing radiation energy-transfer efficiency and photocatalytic activity, resulting in efficient electron–hole pair separation and reactive oxygen species production. After further modification with cancer cell targeting peptide, the obtained nanoplatform demonstrated good performance in U87MG cell uptakes and intracellular radicals-generation, severely damaging the vital subcellular organs of U87MG cells, such as mitochondrion, membrane lipid, and nuclei etc. These combined therapeutic actions mediated by the composition-tunable nanosensitizers significantly inhibited the U87MG tumor growth, providing a refreshing strategy for X-ray induced dynamic therapy of malignant tumors.
Collapse
Affiliation(s)
- Lei Chen
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Jinghui Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, People's Republic of China.,State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts & Telecommunications, Nanjing, 210023, People's Republic of China
| | - Lihua Xu
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Luchao Zhu
- Department of Radiation Oncology, Cancer Center, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, People's Republic of China
| | - Jinpeng Jing
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Yushuo Feng
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Zongzhang Wang
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Peifei Liu
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, People's Republic of China
| | - Wenjing Sun
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, People's Republic of China. .,ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311200, People's Republic of China.
| | - Xiangmei Liu
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts & Telecommunications, Nanjing, 210023, People's Republic of China.
| | - Yimin Li
- Department of Radiation Oncology, Cancer Center, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, People's Republic of China. .,The Third Clinical Medical College, Fujian Medical University, Fuzhou, People's Republic of China.
| | - Hongmin Chen
- State Key Laboratory of Molecular Vaccinology and Molecular, Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, People's Republic of China. .,State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts & Telecommunications, Nanjing, 210023, People's Republic of China. .,Department of Radiation Oncology, Cancer Center, The First Affiliated Hospital of Xiamen University, Xiamen, 361003, People's Republic of China.
| |
Collapse
|
40
|
VİTHANAGE V, C.D. J, M.D.P. DE. C, RAJENDRAM S. Photodynamic Therapy : An Overview and Insights into a Prospective Mainstream Anticancer Therapy. JOURNAL OF THE TURKISH CHEMICAL SOCIETY, SECTION A: CHEMISTRY 2022. [DOI: 10.18596/jotcsa.1000980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Photodynamic therapy (PDT) procedure has minimum invasiveness in contrast to conventional anticancer surgical procedures. Although clinically approved a few decades ago, it is not commonly used due to its poor efficacy, mainly due to poor light penetration into deeper tissues. PDT uses a photosensitizer (PS), which is photoactivated on illumination by light of appropriate wavelength and oxygen in the tissue, leading to a series of photochemical reactions producing reactive oxygen species (ROS) triggering various mechanisms resulting in lethal effects on tumor cells. This review looks into the fundamental aspects of PDT, such as photochemistry, photobiological effects, and the current clinical applications in the light of improving PDT to become a mainstream therapeutic procedure against a broad spectrum of cancers and malignant lesions. The side effects of PDT, both early and late-onset, are elaborated on in detail to highlight the available options to minimize side effects without compromising therapeutic efficacy. This paper summarizes the benefits, drawbacks, and limitations of photodynamic therapy along with the recent attempts to achieve improved therapeutic efficacy via monitoring various cellular and molecular processes through fluorescent imagery aided by suitable biomarkers, prospective nanotechnology-based targeted delivery methods, the use of scintillating nanoparticles to deliver light to remote locations and also combining PDT with conventional anticancer therapies have opened up new dimensions for PDT in treating cancers. This review inquires and critically analyses prospective avenues in which a breakthrough would finally enable PDT to be integrated into mainstream anticancer therapy.
Collapse
|
41
|
Lv R, Raab M, Wang Y, Tian J, Lin J, Prasad PN. Nanochemistry advancing photon conversion in rare-earth nanostructures for theranostics. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214486] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
42
|
Ma Y, Xiao F, Lu C, Wen L. Multifunctional Nanosystems Powered Photodynamic Immunotherapy. Front Pharmacol 2022; 13:905078. [PMID: 35645842 PMCID: PMC9130658 DOI: 10.3389/fphar.2022.905078] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 04/25/2022] [Indexed: 12/19/2022] Open
Abstract
Photodynamic Therapy (PDT) with the intrinsic advantages including non-invasiveness, spatiotemporal selectivity, low side-effects, and immune activation ability has been clinically approved for the treatment of head and neck cancer, esophageal cancer, pancreatic cancer, prostate cancer, and esophageal squamous cell carcinoma. Nevertheless, the PDT is only a strategy for local control of primary tumor, that it is hard to remove the residual tumor cells and inhibit the tumor metastasis. Recently, various smart nanomedicine-based strategies are developed to overcome the barriers of traditional PDT including the drawbacks of traditional photosensitizers, limited tissue penetrability of light, inefficient induction of tumor cell death and tumor resistance to the therapy. More notably, a growing number of studies have focused on improving the therapeutic efficiency by eliciting host immune system with versatile nanoplatforms, which heralds a broader clinical application prospect of PDT in the future. Herein, the pathways of PDT induced-tumor destruction, especially the host immune response is summarized, and focusing on the recent progress of nanosystems-enhanced PDT through eliciting innate immunity and adaptive immunity. We expect it will provide some insights for conquering the drawbacks current PDT and expand the range of clinical application through this review.
Collapse
Affiliation(s)
- Yunong Ma
- Medical College, Guangxi University, Nanning, China
- Zhuhai Precision Medical Center, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated With Jinan University), Jinan University, Zhuhai, China
| | - Fengfeng Xiao
- Zhuhai Precision Medical Center, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated With Jinan University), Jinan University, Zhuhai, China
| | - Cuixia Lu
- Medical College, Guangxi University, Nanning, China
- *Correspondence: Cuixia Lu, ; Liewei Wen,
| | - Liewei Wen
- Zhuhai Precision Medical Center, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated With Jinan University), Jinan University, Zhuhai, China
- *Correspondence: Cuixia Lu, ; Liewei Wen,
| |
Collapse
|
43
|
Deep-Tissue Activation of Photonanomedicines: An Update and Clinical Perspectives. Cancers (Basel) 2022; 14:cancers14082004. [PMID: 35454910 PMCID: PMC9032169 DOI: 10.3390/cancers14082004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Photodynamic therapy (PDT) is a light-activated treatment modality, which is being clinically used and further developed for a number of premalignancies, solid tumors, and disseminated cancers. Nanomedicines that facilitate PDT (photonanomedicines, PNMs) have transformed its safety, efficacy, and capacity for multifunctionality. This review focuses on the state of the art in deep-tissue activation technologies for PNMs and explores how their preclinical use can evolve towards clinical translation by harnessing current clinically available instrumentation. Abstract With the continued development of nanomaterials over the past two decades, specialized photonanomedicines (light-activable nanomedicines, PNMs) have evolved to become excitable by alternative energy sources that typically penetrate tissue deeper than visible light. These sources include electromagnetic radiation lying outside the visible near-infrared spectrum, high energy particles, and acoustic waves, amongst others. Various direct activation mechanisms have leveraged unique facets of specialized nanomaterials, such as upconversion, scintillation, and radiosensitization, as well as several others, in order to activate PNMs. Other indirect activation mechanisms have leveraged the effect of the interaction of deeply penetrating energy sources with tissue in order to activate proximal PNMs. These indirect mechanisms include sonoluminescence and Cerenkov radiation. Such direct and indirect deep-tissue activation has been explored extensively in the preclinical setting to facilitate deep-tissue anticancer photodynamic therapy (PDT); however, clinical translation of these approaches is yet to be explored. This review provides a summary of the state of the art in deep-tissue excitation of PNMs and explores the translatability of such excitation mechanisms towards their clinical adoption. A special emphasis is placed on how current clinical instrumentation can be repurposed to achieve deep-tissue PDT with the mechanisms discussed in this review, thereby further expediting the translation of these highly promising strategies.
Collapse
|
44
|
Ni K, Xu Z, Culbert A, Luo T, Guo N, Yang K, Pearson E, Preusser B, Wu T, La Riviere P, Weichselbaum RR, Spiotto MT, Lin W. Synergistic checkpoint-blockade and radiotherapy–radiodynamic therapy via an immunomodulatory nanoscale metal–organic framework. Nat Biomed Eng 2022; 6:144-156. [DOI: 10.1038/s41551-022-00846-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 01/12/2022] [Indexed: 12/20/2022]
|
45
|
Kang Y, Li Z, Lu F, Su Z, Ji X, Zhang S. Synthesis of red/black phosphorus-based composite nanosheets with a Z-scheme heterostructure for high-performance cancer phototherapy. NANOSCALE 2022; 14:766-779. [PMID: 34951432 DOI: 10.1039/d1nr07553e] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Two dimensional black phosphorus nanosheets (BP NSs) have attracted plenty of attention in the research field of cancer photonic therapy. However, the poor stability and relatively low efficiency of reactive oxygen species (ROS) generation of BP NSs limit their practical application. To address these drawbacks, herein we report a red/black phosphorus (RP/BP) composite nanosheet, M-RP/BP@ZnFe2O4, which was synthesized by (1) partially converting red phosphorus (RP) to black phosphorus (BP) followed by liquid-phase ultrasonic exfoliation to form RP/BP NSs, (2) in situ synthesis of ZnFe2O4 nanoparticles on the surface of RP/BP NSs, (3) and wrapping with the MCF-7 cell membrane. Due to the presence of RP, BP, ZnFe2O4 and the cell membrane, the M-RP/BP@ZnFe2O4 NSs exhibited high performance in cancer phototherapy with the following features: (i) a Z-scheme heterojunction structure was formed between RP/BP NSs thus enabling high separation efficiency of the photogenerated electrons and holes; (ii) the photoexcitation holes in the valence band of RP can break the tumor microenvironment by oxidizing glutathione; (iii) the NSs could decompose water to produce H2O2 and O2, which can be further converted to toxic ˙OH through the ZnFe2O4 catalyzed Fenton reaction and 1O2 through energy transfer, respectively; and (iv) the cell membrane wrapping improved the targeting of the composite NSs at the tumor site and photonic therapy can be finally triggered by a 660 nm laser to convert O2 to ˙O2- and 1O2. The in vitro cytotoxicity experiments showed that more than 90% cells were killed after photodynamic therapy (PDT) at 0.3 mg mL-1 M-RP/BP@ZnFe2O4 NSs, and the animal experiments with xenograft tumor model mice indicated that tumor growth was completely inhibited and the highest survival rate of 83.3% at 60 days post PDT was obtained.
Collapse
Affiliation(s)
- Yong Kang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
- University of Chinese Academy of Sciences, No. 19 Yuquan Road, Shijingshan District, Beijing, 100049, China
| | - Zhengjun Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| | - Fengying Lu
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| | - Zhiguo Su
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| | - Xiaoyuan Ji
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, 300072, China
| | - Songping Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China.
| |
Collapse
|
46
|
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: 2.5] [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
|
47
|
Zheng B, Fan J, Chen B, Qin X, Wang J, Wang F, Deng R, Liu X. Rare-Earth Doping in Nanostructured Inorganic Materials. Chem Rev 2022; 122:5519-5603. [PMID: 34989556 DOI: 10.1021/acs.chemrev.1c00644] [Citation(s) in RCA: 184] [Impact Index Per Article: 92.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Impurity doping is a promising method to impart new properties to various materials. Due to their unique optical, magnetic, and electrical properties, rare-earth ions have been extensively explored as active dopants in inorganic crystal lattices since the 18th century. Rare-earth doping can alter the crystallographic phase, morphology, and size, leading to tunable optical responses of doped nanomaterials. Moreover, rare-earth doping can control the ultimate electronic and catalytic performance of doped nanomaterials in a tunable and scalable manner, enabling significant improvements in energy harvesting and conversion. A better understanding of the critical role of rare-earth doping is a prerequisite for the development of an extensive repertoire of functional nanomaterials for practical applications. In this review, we highlight recent advances in rare-earth doping in inorganic nanomaterials and the associated applications in many fields. This review covers the key criteria for rare-earth doping, including basic electronic structures, lattice environments, and doping strategies, as well as fundamental design principles that enhance the electrical, optical, catalytic, and magnetic properties of the material. We also discuss future research directions and challenges in controlling rare-earth doping for new applications.
Collapse
Affiliation(s)
- Bingzhu Zheng
- State Key Laboratory of Silicon Materials, Institute for Composites Science Innovation, School of Materials Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jingyue Fan
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Bing Chen
- Department of Materials Science and Engineering, City University of Hong Kong, Hong Kong SAR 999077, China
| | - Xian Qin
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Juan Wang
- Institute of Environmental Health, MOE Key Laboratory of Environmental Remediation and Ecosystem Health, College of Environmental & Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Feng Wang
- Department of Materials Science and Engineering, City University of Hong Kong, Hong Kong SAR 999077, China
| | - Renren Deng
- State Key Laboratory of Silicon Materials, Institute for Composites Science Innovation, School of Materials Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Xiaogang Liu
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| |
Collapse
|
48
|
Tiwari AK, Mishra A, Pandey G, Gupta MK, Pandey PC. Nanotechnology: A Potential Weapon to Fight against COVID-19. PARTICLE & PARTICLE SYSTEMS CHARACTERIZATION : MEASUREMENT AND DESCRIPTION OF PARTICLE PROPERTIES AND BEHAVIOR IN POWDERS AND OTHER DISPERSE SYSTEMS 2022; 39:2100159. [PMID: 35440846 PMCID: PMC9011707 DOI: 10.1002/ppsc.202100159] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/28/2021] [Indexed: 05/13/2023]
Abstract
The COVID-19 infections have posed an unprecedented global health emergency, with nearly three million deaths to date, and have caused substantial economic loss globally. Hence, an urgent exploration of effective and safe diagnostic/therapeutic approaches for minimizing the threat of this highly pathogenic coronavirus infection is needed. As an alternative to conventional diagnosis and antiviral agents, nanomaterials have a great potential to cope with the current or even future health emergency situation with a wide range of applications. Fundamentally, nanomaterials are physically and chemically tunable and can be employed for the next generation nanomaterial-based detection of viral antigens and host antibodies in body fluids as antiviral agents, nanovaccine, suppressant of cytokine storm, nanocarrier for efficient delivery of antiviral drugs at infection site or inside the host cells, and can also be a significant tool for better understanding of the gut microbiome and SARS-CoV-2 interaction. The applicability of nanomaterial-based therapeutic options to cope with the current and possible future pandemic is discussed here.
Collapse
Affiliation(s)
- Atul K. Tiwari
- Department of ChemistryIndian Institute of Technology (BHU)VaranasiUttar Pradesh221005India
| | - Anupa Mishra
- Department of MicrobiologyDr. R.M.L. Awadh UniversityAyodhyaUttar Pradesh224001India
- Department of MicrobiologySri Raghukul Mahila Vidya PeethCivil Line GondaUttar Pradesh271001India
| | - Govind Pandey
- Department of PaediatricsKing George Medical UniversityLucknowUttar Pradesh226003India
| | - Munesh K. Gupta
- Department of MicrobiologyInstitute of Medical SciencesBanaras Hindu UniversityVaranasiUttar Pradesh221005India
| | - Prem C. Pandey
- Department of ChemistryIndian Institute of Technology (BHU)VaranasiUttar Pradesh221005India
| |
Collapse
|
49
|
Wang C, Wang H, Luo L, Gan S, Yao Y, Wei Q, Wu J, Yuan A, Hu Y, Wu C, Zou Z. Scintillator-based radiocatalytic superoxide radical production for long-term tumor DNA damage. Biomater Sci 2022; 10:3433-3440. [DOI: 10.1039/d2bm00101b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Photocatalytic materials absorb photons ranging from ultraviolet to near-infrared light to initiate photocatalytic reactions and have broad application prospects in various fields. However, high-energy ionizing radiations are rarely involved in...
Collapse
|
50
|
Abstract
Photodynamic therapy (PDT) is an emerging treatment option for cancer. In PDT, photosensitizers are delivered to tumors and stimulated by light to generate reactive oxygen species (ROS)-most importantly singlet oxygen (1O2)-to damage tumor cells or induce tissue ischemia. PDT is associated with a low level of systemic toxicity because photosensitizers are usually pharmaceutically inactive in the dark and photoirradiation is applied only to tumor areas in the procedure. Additionally, PDT can be applied repeatedly without cumulative toxicity or incurring resistance, and may stimulate systemic anti-tumor immunity. However, PDT's clinical use has been restricted due to the limited penetration of visible light through tissues. X-rays possess superior tissue penetration capability and are exploited in X-ray-induced photodynamic therapy to overcome this limitation. Herein we have demonstrated this principle with a novel LiGa5O8:Cr (LGO:Cr)-based nanoscintillator which emits near-infrared X-ray luminescence to both guide external beam therapy and induce PDT with the photosensitizer (2,3-naphthalocyanine) encapsulated in a mesoporous silica shell of the nanoscintillator.
Collapse
Affiliation(s)
- Benjamin Cline
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Jin Xie
- Department of Chemistry, University of Georgia, Athens, GA, USA.
| |
Collapse
|