1
|
Lee WS, Kokubo T, Choi Y, Hamano T, Zaboronok A, Ishikawa T, Kwon OD, Kim E, Kim JK. Carbon ion stimulation therapy reverses iron deposits and microglia driven neuroinflammation and induces cognitive improvement in an Alzheimer's disease mouse model. Sci Rep 2025; 15:7938. [PMID: 40050677 PMCID: PMC11885615 DOI: 10.1038/s41598-025-91689-3] [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: 08/22/2024] [Accepted: 02/21/2025] [Indexed: 03/09/2025] Open
Abstract
Insoluble iron deposits often exist as iron oxide nanoparticles in protein aggregates, impaired ferritin, or activated microglia and have been implicated as major causes of neuroinflammation in Alzheimer's disease. However, no crucial evidence has been reported to support the therapeutic effects of current iron chelators on the deposition of various molecular forms of insoluble iron. We investigated the therapeutic effect of carbon ion stimulation (CIS) via a transmission beam on insoluble iron deposits, iron inclusion bodies, and the associated biological response in 5xFAD AD mouse brains. Compared with no treatment, CIS dose-dependently induced a 33-60% reduction in the amount of ferrous-containing iron species and associated inclusion bodies in the brains of AD mice. CIS induced considerable neuroinflammation downregulation and, conversely, anti-inflammatory upregulation, which was associated with improved memory and enhanced hippocampal neurogenesis. In conclusion, our results suggest that the effective degradation of insoluble iron deposits in combination with pathogenic inclusion bodies promotes AD-modifying properties and offers a potential CIS treatment option for AD.
Collapse
Affiliation(s)
- Won-Seok Lee
- Department of Biochemistry, Daegu Catholic University, Daegu, 42472, South Korea
| | | | - Younshick Choi
- Department of a Biomedical Engineering & Radiology, Daegu Catholic University, Daegu, 42472, South Korea
| | | | - Alexander Zaboronok
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Takaaki Ishikawa
- Department of Neurosurgery, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Oh-Dae Kwon
- Department of Neurology, School of Medicine, Daegu Catholic University, Daegu, 42472, South Korea
| | - EunHo Kim
- Department of Biochemistry, Daegu Catholic University, Daegu, 42472, South Korea.
| | - Jong-Ki Kim
- Department of a Biomedical Engineering & Radiology, Daegu Catholic University, Daegu, 42472, South Korea.
| |
Collapse
|
2
|
Patel KD, Keskin-Erdogan Z, Sawadkar P, Nik Sharifulden NSA, Shannon MR, Patel M, Silva LB, Patel R, Chau DYS, Knowles JC, Perriman AW, Kim HW. Oxidative stress modulating nanomaterials and their biochemical roles in nanomedicine. NANOSCALE HORIZONS 2024; 9:1630-1682. [PMID: 39018043 DOI: 10.1039/d4nh00171k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Many pathological conditions are predominantly associated with oxidative stress, arising from reactive oxygen species (ROS); therefore, the modulation of redox activities has been a key strategy to restore normal tissue functions. Current approaches involve establishing a favorable cellular redox environment through the administration of therapeutic drugs and redox-active nanomaterials (RANs). In particular, RANs not only provide a stable and reliable means of therapeutic delivery but also possess the capacity to finely tune various interconnected components, including radicals, enzymes, proteins, transcription factors, and metabolites. Here, we discuss the roles that engineered RANs play in a spectrum of pathological conditions, such as cancer, neurodegenerative diseases, infections, and inflammation. We visualize the dual functions of RANs as both generator and scavenger of ROS, emphasizing their profound impact on diverse cellular functions. The focus of this review is solely on inorganic redox-active nanomaterials (inorganic RANs). Additionally, we deliberate on the challenges associated with current RANs-based approaches and propose potential research directions for their future clinical translation.
Collapse
Affiliation(s)
- Kapil D Patel
- John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia.
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
- School of Cellular and Molecular Medicine, University of Bristol, BS8 1TD, UK
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea.
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
| | - Zalike Keskin-Erdogan
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, Royal Free Hospital, Rowland Hill Street, NW3 2PF, London, UK
- Department of Chemical Engineering, Imperial College London, Exhibition Rd, South Kensington, SW7 2BX, London, UK
| | - Prasad Sawadkar
- Division of Surgery and Interventional Science, UCL, London, UK
- The Griffin Institute, Northwick Park Institute for Medical Research, Northwick Park and St Mark's Hospitals, London, HA1 3UJ, UK
| | - Nik Syahirah Aliaa Nik Sharifulden
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, Royal Free Hospital, Rowland Hill Street, NW3 2PF, London, UK
| | - Mark Robert Shannon
- John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia.
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
- School of Cellular and Molecular Medicine, University of Bristol, BS8 1TD, UK
| | - Madhumita Patel
- Department of Chemistry and Nanoscience, Ewha Women University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Republic of Korea
| | - Lady Barrios Silva
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, Royal Free Hospital, Rowland Hill Street, NW3 2PF, London, UK
| | - Rajkumar Patel
- Energy & Environment Sciences and Engineering (EESE), Integrated Sciences and Engineering Division (ISED), Underwood International College, Yonsei University, 85 Songdongwahak-ro, Yeonsungu, Incheon 21938, Republic of Korea
| | - David Y S Chau
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, Royal Free Hospital, Rowland Hill Street, NW3 2PF, London, UK
| | - Jonathan C Knowles
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, Royal Free Hospital, Rowland Hill Street, NW3 2PF, London, UK
| | - Adam W Perriman
- John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia.
- Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia
- School of Cellular and Molecular Medicine, University of Bristol, BS8 1TD, UK
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea.
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
3
|
Yu B, Lu X, Feng X, Zhao T, Li J, Lu Y, Ye F, Liu X, Zheng X, Shen Z, Jin X, Chen W, Li Q. Gadolinium Oxide Nanoparticles Reinforce the Fractionated Radiotherapy-Induced Immune Response in Tri-Negative Breast Cancer via cGAS-STING Pathway. Int J Nanomedicine 2023; 18:7713-7728. [PMID: 38115988 PMCID: PMC10729773 DOI: 10.2147/ijn.s428044] [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: 08/20/2023] [Accepted: 12/02/2023] [Indexed: 12/21/2023] Open
Abstract
Introduction Radiotherapy is a widely recognized first-line clinical treatment for cancer, but its efficacy may be impeded by the radioresistance of advanced tumors. It is urgent to improve the sensitivity of radioresistant tumors to radiotherapy. In this work, gadolinium oxide nanocrystals (GONs) were utilized as radiosensitizers to enhance the killing effect and reinforce the immune activation of X-ray irradiation on 4T1 breast cancer cells in vitro and in vivo. Methods 1.0 T small animal MR imaging (MRI) system was employed to trace GONs in vivo, while 225 kVp X-ray irradiation equipment was utilized for investigating the radiosensitization of GONs in 4T1 breast cancer cells in vitro and in vivo. Western blot, quantitative real-time PCR (RT-qPCR), immunohistochemistry, immunofluorescence, clonal survival assay, flow cytometry and reactive oxygen species assay were used to explore the biological mechanism of GON sensitization. Results GONs exhibited exceptional utility as contrast agents for both in vivo and in vitro MRI imaging. Interestingly, a single dose of 8.0 Gy X-rays together with GONs failed to confer superior therapeutic effects in tumor-bearing mice, while only 3.0 Gy × 3 fractions X-rays combined with GONs exhibited effective tumor growth inhibition. Moreover, fractionated X-ray irradiation with GONs demonstrated a superior capacity to activate the cGAS-STING pathway. Discussion Fractionated X-ray irradiation in the presence of GONs has demonstrated the most significant activation of the anti-tumor immune response by boosting the cGAS-STING pathway.
Collapse
Affiliation(s)
- Boyi Yu
- Biomedical Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, People’s Republic of China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, People’s Republic of China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province, People’s Republic of China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Xuanyi Lu
- School of Biomedical Engineering, Southern Medical University, Guangzhou, People’s Republic of China
| | - Xianglong Feng
- Biomedical Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, People’s Republic of China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, People’s Republic of China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province, People’s Republic of China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Ting Zhao
- Biomedical Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, People’s Republic of China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, People’s Republic of China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province, People’s Republic of China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Jiaxin Li
- Biomedical Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, People’s Republic of China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, People’s Republic of China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province, People’s Republic of China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Yudie Lu
- School of Biomedical Engineering, Southern Medical University, Guangzhou, People’s Republic of China
| | - Fei Ye
- Biomedical Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, People’s Republic of China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, People’s Republic of China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province, People’s Republic of China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Xiongxiong Liu
- Biomedical Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, People’s Republic of China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, People’s Republic of China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province, People’s Republic of China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Xiaogang Zheng
- Biomedical Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, People’s Republic of China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, People’s Republic of China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province, People’s Republic of China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Zheyu Shen
- School of Biomedical Engineering, Southern Medical University, Guangzhou, People’s Republic of China
| | - Xiaodong Jin
- Biomedical Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, People’s Republic of China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, People’s Republic of China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province, People’s Republic of China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Weiqiang Chen
- Biomedical Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, People’s Republic of China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, People’s Republic of China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province, People’s Republic of China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Qiang Li
- Biomedical Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, People’s Republic of China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine, Chinese Academy of Sciences, Lanzhou, People’s Republic of China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Lanzhou, Gansu Province, People’s Republic of China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| |
Collapse
|
4
|
Chen SF, Kau M, Wang YC, Chen MH, Tung FI, Chen MH, Liu TY. Synergistically Enhancing Immunotherapy Efficacy in Glioblastoma with Gold-Core Silica-Shell Nanoparticles and Radiation. Int J Nanomedicine 2023; 18:7677-7693. [PMID: 38111846 PMCID: PMC10726961 DOI: 10.2147/ijn.s440405] [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: 09/16/2023] [Accepted: 12/12/2023] [Indexed: 12/20/2023] Open
Abstract
Purpose Glioblastoma is a highly aggressive brain tumor with universally poor outcomes. Recent progress in immune checkpoint inhibitors has led to increased interest in their application in glioblastoma. Nonetheless, the unique immune milieu in the brain has posed remarkable challenges to the efficacy of immunotherapy. We aimed to leverage the radiation-induced immunogenic cell death to overcome the immunosuppressive network in glioblastoma. Methods We developed a novel approach using the gold-core silica-shell nanoparticles (Au@SiO2 NPs) in combination with low-dose radiation to enhance the therapeutic efficacy of the immune checkpoint inhibitor (atezolizumab) in brain tumors. The biocompatibility, immune cell recruitment, and antitumor ability of the combinatorial strategy were determined using in vitro assays and in vivo models. Results Our approach successfully induced the migration of macrophages towards brain tumors and promoted cancer cell apoptosis. Subcutaneous tumor models demonstrated favorable safety profiles and significantly enhanced anticancer effects. In orthotopic brain tumor models, the multimodal therapy yielded substantial prognostic benefits over any individual modalities, achieving an impressive 40% survival rate. Conclusion In summary, the combination of Au@SiO2 NPs and low-dose radiation holds the potential to improve the clinical efficacy of immune checkpoint inhibitors. The synergetic strategy modulates tumor microenvironments and enhances systemic antitumor immunity, paving a novel way for glioblastoma treatment.
Collapse
Affiliation(s)
- Shuo-Fu Chen
- Department of Heavy Particles & Radiation Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Min Kau
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Chi Wang
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ming-Hong Chen
- Division of Neurosurgery, Department of Surgery, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Fu-I Tung
- Department of Orthopedics, Yang-Ming Branch, Taipei City Hospital, Taipei, Taiwan
- Department of Health and Welfare, College of City Management, University of Taipei, Taipei, Taiwan
| | - Mei-Hsiu Chen
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Tse-Ying Liu
- Department of Biomedical Engineering, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
5
|
Gerken LRH, Gerdes ME, Pruschy M, Herrmann IK. Prospects of nanoparticle-based radioenhancement for radiotherapy. MATERIALS HORIZONS 2023; 10:4059-4082. [PMID: 37555747 PMCID: PMC10544071 DOI: 10.1039/d3mh00265a] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 08/02/2023] [Indexed: 08/10/2023]
Abstract
Radiotherapy is a key pillar of solid cancer treatment. Despite a high level of conformal dose deposition, radiotherapy is limited due to co-irradiation of organs at risk and subsequent normal tissue toxicities. Nanotechnology offers an attractive opportunity for increasing the efficacy and safety of cancer radiotherapy. Leveraging the freedom of design and the growing synthetic capabilities of the nanomaterial-community, a variety of engineered nanomaterials have been designed and investigated as radiosensitizers or radioenhancers. While research so far has been primarily focused on gold nanoparticles and other high atomic number materials to increase the absorption cross section of tumor tissue, recent studies are challenging the traditional concept of high-Z nanoparticle radioenhancers and highlight the importance of catalytic activity. This review provides a concise overview on the knowledge of nanoparticle radioenhancement mechanisms and their quantification. It critically discusses potential radioenhancer candidate materials and general design criteria for different radiation therapy modalities, and concludes with research priorities in order to advance the development of nanomaterials, to enhance the efficacy of radiotherapy and to increase at the same time the therapeutic window.
Collapse
Affiliation(s)
- Lukas R H Gerken
- Nanoparticle Systems Engineering Laboratory, Institute of Energy and Process Engineering (IEPE), Department of Mechanical and Process Engineering (D-MAVT), ETH Zurich, Sonneggstrasse 3, 8092 Zurich, Switzerland.
- Particles-Biology Interactions Laboratory, Department of Materials Meet Life, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| | - Maren E Gerdes
- Karolinska Institutet, Solnavägen 1, 171 77 Stockholm, Sweden
| | - Martin Pruschy
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Inge K Herrmann
- Nanoparticle Systems Engineering Laboratory, Institute of Energy and Process Engineering (IEPE), Department of Mechanical and Process Engineering (D-MAVT), ETH Zurich, Sonneggstrasse 3, 8092 Zurich, Switzerland.
- Particles-Biology Interactions Laboratory, Department of Materials Meet Life, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, 9014 St. Gallen, Switzerland
| |
Collapse
|
6
|
Sisin NNT, Rahman WN. Potentials of Bismuth-Based Nanoparticles and Baicalein Natural Compounds as Radiosensitizers in Cancer Radiotherapy: a Review. BIONANOSCIENCE 2023. [DOI: 10.1007/s12668-022-01057-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
7
|
Low-Energy Electron Generation for Biomolecular Damage Inquiry: Instrumentation and Methods. BIOPHYSICA 2022. [DOI: 10.3390/biophysica2040041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Technological advancement has produced a variety of instruments and methods to generate electron beams that have greatly assisted in the extensive theoretical and experimental efforts devoted to investigating the effect of secondary electrons with energies approximately less than 100 eV, which are referred as low-energy electrons (LEEs). In the past two decades, LEE studies have focused on biomolecular systems, which mainly consist of DNA and proteins and their constituents as primary cellular targets of ionizing radiation. These studies have revealed that compared to other reactive species produced by high-energy radiation, LEEs have distinctive pathways and considerable efficiency in inducing lethal DNA lesions. The present work aims to briefly discuss the current state of LEE production technology and to motivate further studies and improvements of LEE generation techniques in relation to biological electron-driven processes associated with such medical applications as radiation therapy and cancer treatment.
Collapse
|
8
|
Mesbahi A, Rajabpour S, Smilowitz HM, Hainfeld JF. Accelerated brachytherapy with the Xoft electronic source used in association with iodine, gold, bismuth, gadolinium, and hafnium nano-radioenhancers. Brachytherapy 2022; 21:968-978. [PMID: 36002350 DOI: 10.1016/j.brachy.2022.06.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/07/2022] [Accepted: 06/30/2022] [Indexed: 02/07/2023]
Abstract
PURPOSE The current study was designed to calculate the dose enhancement factor (DEF) of iodine (I), gold (Au), bismuth (Bi), gadolinium (Gd), and hafnium (Hf) nanoparticles (NP)s by Monte Carlo (MC) modeling of an electronic brachytherapy source in resection cavities of breast tumors. METHODS AND MATERIALS The GEANT4 MC code was used for simulation of a phantom containing a water-filled balloon and a Xoft source (50 kVp) to irradiate the margins of a resected breast tumor. NPs with a diameter of 20 nm and concentrations from 1 to 5% w/w were simulated in a tumor margin with 5 mm thickness as well as a hypothetical breast model consisting of spherical island-like residual tumor-remnants. The DEFs for all NPs were calculated in both models. RESULTS In the margin-loaded model, for the concentration of 1% w/w heavy atom, DEFs of 2.5, 2.3, 2.1, 2, and 1.7 were calculated for Bi, Au, I, Hf, and Gd NPs (descending order), which increased, almost linearly with concentration for all NPs. Moreover, normal tissue dose behind the NP-loaded margin declined significantly depending on NP type and concentration. When modeling residual tumor islands, DEF values were very close to the margin-loaded values except for Bi and I, where DEFs of 2.55 and 1.7 were seen, respectively. CONCLUSIONS Considerable dose enhancements were obtained for the heavy atom NPs studied in the partial breast brachytherapy with a Xoft electronic source. In addition, normal tissue doses were lowered in the points beyond the NP-loaded margin. The findings revealed promising outcomes and the probability of improved tumor control for NP-aided brachytherapy with the Xoft electronic source.
Collapse
Affiliation(s)
- Asghar Mesbahi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Medical Radiation Sciences Research Team, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Saeed Rajabpour
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Henry M Smilowitz
- Department of Cell Biology, University of Connecticut Health Center, CT
| | | |
Collapse
|
9
|
Higashi Y, Ma Y, Matsumoto K, Shiro A, Saitoh H, Kawachi T, Tamanoi F. Auger electrons and DNA double-strand breaks studied by using iodine-containing chemicals. Enzymes 2022; 51:101-115. [PMID: 36336404 DOI: 10.1016/bs.enz.2022.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Irradiation of high Z elements such as iodine, gold, gadolinium with monochromatic X-rays causes photoelectric effects that include the release of Auger electrons. Decay of radioactive iodine such as I-123 and I-125 also results in multiple events and some involve the generation of Auger electrons. These electrons have low energy and travel only a short distance but have a strong effect on DNA damage including the generation of double-strand breaks. In this chapter, we focus on iodine and discuss various studies that used iodine-containing chemicals to generate Auger electrons and cause DNA double-strand breaks. First, DNA synthesis precursors containing iodine were used to place iodine on DNA. DNA binding dyes such as iodine Hoechst were investigated for Auger electron generation and DNA breaks. More recently, iodine containing nanoparticles were developed. We describe our study using tumor spheroids loaded with iodine nanoparticles and synchrotron-generated monochromatic X-rays. This study led to the demonstration that an optimum effect on DNA double-strand break formation is observed with a 33.2keV X-ray which is just above the K-edge energy of iodine.
Collapse
Affiliation(s)
- Yuya Higashi
- Institute for Integrated Cell-Material Sciences, Institute for Advanced Study, Kyoto University, Kyoto, Japan
| | - Yue Ma
- Institute for Integrated Cell-Material Sciences, Institute for Advanced Study, Kyoto University, Kyoto, Japan; Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Kotaro Matsumoto
- Institute for Integrated Cell-Material Sciences, Institute for Advanced Study, Kyoto University, Kyoto, Japan
| | - Ayumi Shiro
- Kansai Photon Science Institute, Quantum Beam Science Research Directorate, National Institutes for Quantum Science and Technology, Hyogo, Japan
| | - Hiroyuki Saitoh
- Kansai Photon Science Institute, Quantum Beam Science Research Directorate, National Institutes for Quantum Science and Technology, Hyogo, Japan
| | - Tetsuya Kawachi
- Kansai Photon Science Institute, Quantum Beam Science Research Directorate, National Institute for Quantum Science and Technology, Kizu, Japan
| | - Fuyuhiko Tamanoi
- Institute for Integrated Cell-Material Sciences, Institute for Advanced Study, Kyoto University, Kyoto, Japan
| |
Collapse
|
10
|
Iodine containing porous organosilica nanoparticles trigger tumor spheroids destruction upon monochromatic X-ray irradiation: DNA breaks and K-edge energy X-ray. Sci Rep 2021; 11:14192. [PMID: 34262055 PMCID: PMC8280225 DOI: 10.1038/s41598-021-93429-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/17/2021] [Indexed: 02/06/2023] Open
Abstract
X-ray irradiation of high Z elements causes photoelectric effects that include the release of Auger electrons that can induce localized DNA breaks. We have previously established a tumor spheroid-based assay that used gadolinium containing mesoporous silica nanoparticles and synchrotron-generated monochromatic X-rays. In this work, we focused on iodine and synthesized iodine-containing porous organosilica (IPO) nanoparticles. IPO were loaded onto tumor spheroids and the spheroids were irradiated with 33.2 keV monochromatic X-ray. After incubation in CO2 incubator, destruction of tumor spheroids was observed which was accompanied by apoptosis induction, as determined by the TUNEL assay. By employing the γH2AX assay, we detected double strand DNA cleavages immediately after the irradiation. These results suggest that IPO first generate double strand DNA breaks upon X-ray irradiation followed by apoptosis induction of cancer cells. Use of three different monochromatic X-rays having energy levels of 33.0, 33.2 and 33.4 keV as well as X-rays with 0.1 keV energy intervals showed that the optimum effect of all three events (spheroid destruction, apoptosis induction and generation of double strand DNA breaks) occurred with a 33.2 keV monochromatic X-ray. These results uncover the preferential effect of K-edge energy X-ray for tumor spheroid destruction mediated by iodine containing nanoparticles.
Collapse
|
11
|
Tremi I, Spyratou E, Souli M, Efstathopoulos EP, Makropoulou M, Georgakilas AG, Sihver L. Requirements for Designing an Effective Metallic Nanoparticle (NP)-Boosted Radiation Therapy (RT). Cancers (Basel) 2021; 13:cancers13133185. [PMID: 34202342 PMCID: PMC8269428 DOI: 10.3390/cancers13133185] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Recent advances in nanotechnology gave rise to trials with various types of metallic nanoparticles (NPs) to enhance the radiosensitization of cancer cells while reducing or maintaining the normal tissue complication probability during radiation therapy. This work reviews the physical and chemical mechanisms leading to the enhancement of ionizing radiation’s detrimental effects on cells and tissues, as well as the plethora of experimental procedures to study these effects of the so-called “NPs’ radiosensitization”. The paper presents the need to a better understanding of all the phases of actions before applying metallic-based NPs in clinical practice to improve the effect of IR therapy. More physical and biological experiments especially in vivo must be performed and simulation Monte Carlo or mathematical codes based on more accurate models for all phases must be developed. Abstract Many different tumor-targeted strategies are under development worldwide to limit the side effects and improve the effectiveness of cancer therapies. One promising method is to enhance the radiosensitization of the cancer cells while reducing or maintaining the normal tissue complication probability during radiation therapy using metallic nanoparticles (NPs). Radiotherapy with MV photons is more commonly available and applied in cancer clinics than high LET particle radiotherapy, so the addition of high-Z NPs has the potential to further increase the efficacy of photon radiotherapy in terms of NP radiosensitization. Generally, when using X-rays, mainly the inner electron shells are ionized, which creates cascades of both low and high energy Auger electrons. When using high LET particles, mainly the outer shells are ionized, which give electrons with lower energies than when using X-rays. The amount of the produced low energy electrons is higher when exposing NPs to heavy charged particles than when exposing them to X-rays. Since ions traverse the material along tracks, and therefore give rise to a much more inhomogeneous dose distributions than X-rays, there might be a need to introduce a higher number of NPs when using ions compared to when using X-rays to create enough primary and secondary electrons to get the desired dose escalations. This raises the questions of toxicity. This paper provides a review of the fundamental processes controlling the outcome of metallic NP-boosted photon beam and ion beam radiation therapy and presents some experimental procedures to study the biological effects of NPs’ radiosensitization. The overview shows the need for more systematic studies of the behavior of NPs when exposed to different kinds of ionizing radiation before applying metallic-based NPs in clinical practice to improve the effect of IR therapy.
Collapse
Affiliation(s)
- Ioanna Tremi
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, Zografou Campus, National Technical University of Athens (NTUA), 15780 Athens, Greece; (I.T.); (M.S.); (M.M.)
| | - Ellas Spyratou
- 2nd Department of Radiology, Medical School, National and Kapodistrian University of Athens, 11517 Athens, Greece; (E.S.); (E.P.E.)
| | - Maria Souli
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, Zografou Campus, National Technical University of Athens (NTUA), 15780 Athens, Greece; (I.T.); (M.S.); (M.M.)
- Atominstitut, Technische Universität Wien, Stadionallee 2, 1020 Vienna, Austria
| | - Efstathios P. Efstathopoulos
- 2nd Department of Radiology, Medical School, National and Kapodistrian University of Athens, 11517 Athens, Greece; (E.S.); (E.P.E.)
| | - Mersini Makropoulou
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, Zografou Campus, National Technical University of Athens (NTUA), 15780 Athens, Greece; (I.T.); (M.S.); (M.M.)
| | - Alexandros G. Georgakilas
- DNA Damage Laboratory, Department of Physics, School of Applied Mathematical and Physical Sciences, Zografou Campus, National Technical University of Athens (NTUA), 15780 Athens, Greece; (I.T.); (M.S.); (M.M.)
- Correspondence: (A.G.G.); (L.S.)
| | - Lembit Sihver
- Atominstitut, Technische Universität Wien, Stadionallee 2, 1020 Vienna, Austria
- Department of Physics, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden
- Correspondence: (A.G.G.); (L.S.)
| |
Collapse
|
12
|
An in silico study on the effect of host tissue at brachytherapy dose enhancement by gold nanoparticles. Brachytherapy 2020; 20:420-425. [PMID: 33317965 DOI: 10.1016/j.brachy.2020.10.014] [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: 05/15/2020] [Revised: 09/14/2020] [Accepted: 10/17/2020] [Indexed: 11/20/2022]
Abstract
PURPOSE Iridium-192 brachytherapy dose enhancement by gold nanoparticles was investigated in five different tumor tissues to observe the tissue-related differences as an effective environmental factor in the applications of nanoparticles as radio-enhancer agents. METHODS AND MATERIALS The brachytherapy high-dose-rate source of BEBIG Ir-192, a tumor volume with five different tissues including water, Plexiglas, soft tissue, adipose, and bone with and without a uniform distribution of gold nanoparticles were mimicked by MCNPX Monte Carlo simulation code using lattice feature. Dose enhancement factors in the tumor volume were measured separately regarding the types of tissue, and a previous study using GEometry ANd Tracking 4 simulation was used for result validation. RESULTS The results demonstrated that various types of tissue, as the host of gold nanoparticles, lead to different dose enhancement level, so that the bone and adipose have the lowest and the highest amount of dose enhancement factor with values 20.8% and 39.75%, respectively. The maximum difference of 4.8% was achieved from data benchmarking. CONCLUSIONS The results of this study indicate that the MCNPX code can be used as a valid tool for dose measurement in the presence of nanoparticles. Moreover, tissue types of tumor as an environmental feature, alongside with the nanoparticle's size and concentration as well as the conditions of radiotherapy, should be considered in the dose calculation.
Collapse
|
13
|
Monte Carlo characterization of the gold nanoparticles dose enhancement and estimation of the physical interactions weight in dose enhancement mechanism. POLISH JOURNAL OF MEDICAL PHYSICS AND ENGINEERING 2020. [DOI: 10.2478/pjmpe-2020-0026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Abstract
Radiosensitization of the cancer cells by the heavy atoms of nanoparticles was the subject of some studies. But, the physical characterization to determine the weight of all interactions hasn’t been made numerically. The aim of this study was to calculate and compare the dose enhancement (DE) for different energies. The Monte Carlo simulation method was used in the current study. The influence of gold nanoparticles (GNP) size, beam quality, the GNP concentration, and dose inhomogeneity on the radiosensitization by DE was studied. A 35% increase in the photoelectric effect was observed while energy decreased from 18 MV to 300 kV. In the microscopic study which DE calculated in 30 µm from a single GNP, a 79% decreasing in DE within the first 1µm was seen and it declined to 2% in 30 µm from the GNP center. The effect was observed at small distances only. Our study revealed that the dose inhomogeneity around a nanoparticle is the main and very strong effect of DE on a macroscopic scale. In the location which 35% DE occurs most malignant cells survival will be effectively reduced. Our research indicates the need for further research.
Collapse
|
14
|
Jahnke T, Hergenhahn U, Winter B, Dörner R, Frühling U, Demekhin PV, Gokhberg K, Cederbaum LS, Ehresmann A, Knie A, Dreuw A. Interatomic and Intermolecular Coulombic Decay. Chem Rev 2020; 120:11295-11369. [PMID: 33035051 PMCID: PMC7596762 DOI: 10.1021/acs.chemrev.0c00106] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Indexed: 12/11/2022]
Abstract
Interatomic or intermolecular Coulombic decay (ICD) is a nonlocal electronic decay mechanism occurring in weakly bound matter. In an ICD process, energy released by electronic relaxation of an excited atom or molecule leads to ionization of a neighboring one via Coulombic electron interactions. ICD has been predicted theoretically in the mid nineties of the last century, and its existence has been confirmed experimentally approximately ten years later. Since then, a number of fundamental and applied aspects have been studied in this quickly growing field of research. This review provides an introduction to ICD and draws the connection to related energy transfer and ionization processes. The theoretical approaches for the description of ICD as well as the experimental techniques developed and employed for its investigation are described. The existing body of literature on experimental and theoretical studies of ICD processes in different atomic and molecular systems is reviewed.
Collapse
Affiliation(s)
- Till Jahnke
- Institut
für Kernphysik, Goethe Universität, Max-von-Laue-Str. 1, 60438 Frankfurt, Germany
| | - Uwe Hergenhahn
- Fritz-Haber-Institut
der Max-Planck-Gesellschaft, Faradayweg 4-6, 14195 Berlin, Germany
- Max
Planck Institute for Plasma Physics, Wendelsteinstr. 1, 17491 Greifswald, Germany
- Leibniz
Institute of Surface Engineering (IOM), 04318 Leipzig, Germany
| | - Bernd Winter
- Fritz-Haber-Institut
der Max-Planck-Gesellschaft, Faradayweg 4-6, 14195 Berlin, Germany
| | - Reinhard Dörner
- Institut
für Kernphysik, Goethe Universität, Max-von-Laue-Str. 1, 60438 Frankfurt, Germany
| | - Ulrike Frühling
- Institut
für Experimentalphysik and Center for Ultrafast Imaging, Universität Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany
| | - Philipp V. Demekhin
- Institut
für Physik und CINSaT, Universität
Kassel, Heinrich-Plett-Str. 40, 34132 Kassel, Germany
| | - Kirill Gokhberg
- Physical-Chemistry
Institute, Ruprecht-Karls University, Im Neuenheimer Feld 229, 69120 Heidelberg, Germany
| | - Lorenz S. Cederbaum
- Physical-Chemistry
Institute, Ruprecht-Karls University, Im Neuenheimer Feld 229, 69120 Heidelberg, Germany
| | - Arno Ehresmann
- Institut
für Physik und CINSaT, Universität
Kassel, Heinrich-Plett-Str. 40, 34132 Kassel, Germany
| | - André Knie
- Institut
für Physik und CINSaT, Universität
Kassel, Heinrich-Plett-Str. 40, 34132 Kassel, Germany
| | - Andreas Dreuw
- Interdisciplinary
Center for Scientific Computing, Ruprecht-Karls
University, Im Neuenheimer
Feld 205, 69120 Heidelberg, Germany
| |
Collapse
|
15
|
Choi J, Kim G, Cho SB, Im HJ. Radiosensitizing high-Z metal nanoparticles for enhanced radiotherapy of glioblastoma multiforme. J Nanobiotechnology 2020; 18:122. [PMID: 32883290 PMCID: PMC7470617 DOI: 10.1186/s12951-020-00684-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 08/28/2020] [Indexed: 12/13/2022] Open
Abstract
Radiotherapy is an essential step during the treatment of glioblastoma multiforme (GBM), one of the most lethal malignancies. The survival in patients with GBM was improved by the current standard of care for GBM established in 2005 but has stagnated since then. Since GBM is a radioresistant malignancy and the most of GBM recurrences occur in the radiotherapy field, increasing the effectiveness of radiotherapy using high-Z metal nanoparticles (NPs) has recently attracted attention. This review summarizes the progress in radiotherapy approaches for the current treatment of GBM, the physical and biological mechanisms of radiosensitization through high-Z metal NPs, and the results of studies on radiosensitization in the in vitro and in vivo GBM models using high-Z metal NPs to date.
Collapse
Affiliation(s)
- Jinyeong Choi
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Gaeun Kim
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Su Bin Cho
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Hyung-Jun Im
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
16
|
Gadolinium Oxide Nanoparticles Induce Toxicity in Human Endothelial HUVECs via Lipid Peroxidation, Mitochondrial Dysfunction and Autophagy Modulation. NANOMATERIALS 2020; 10:nano10091675. [PMID: 32859033 PMCID: PMC7559735 DOI: 10.3390/nano10091675] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/20/2020] [Accepted: 08/23/2020] [Indexed: 12/31/2022]
Abstract
In spite of the potential preclinical advantage of Gd2O3 nanoparticles (designated here as GO NPs) over gadolinium-based compounds in MRI, recent concerns of gadolinium deposits in various tissues undergoing MRI demands a mechanistic investigation. Hence, we chose human to measure umbilical vein endothelial cells (HUVECs) that line the vasculature and relevant biomarkers due to GO NPs exposure in parallel with the NPs of ZnO as a positive control of toxicity. GO NPs, as measured by TEM, had an average length of 54.8 ± 29 nm and a diameter of 13.7 ± 6 nm suggesting a fiber-like appearance. With not as pronounced toxicity associated with a 24-h exposure, GO NPs induced a concentration-dependent cytotoxicity (IC50 = 304 ± 17 µg/mL) in HUVECs when exposed for 48 h. GO NPs emerged as significant inducer of lipid peroxidation (LPO), reactive oxygen species (ROS), mitochondrial membrane potential (MMP) and autophagic vesicles in comparison to that caused by ZnO NPs at its IC50 for the same exposure time (48 h). While ZnO NPs clearly appeared to induce apoptosis, GO NPs revealed both apoptotic as well as necrotic potentials in HUVECs. Intriguingly, the exogenous antioxidant NAC (N-acetylcysteine) co-treatment significantly attenuated the oxidative imbalance due to NPs preventing cytotoxicity significantly.
Collapse
|
17
|
Barium tungstate nanoparticles to enhance radiation therapy against cancer. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 28:102230. [DOI: 10.1016/j.nano.2020.102230] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 02/29/2020] [Accepted: 05/26/2020] [Indexed: 11/24/2022]
|
18
|
Landry C, Morrison A, Ghandi K. Application of muon and other complementary radiation techniques to study interaction of radiation with nanostructures. Radiat Phys Chem Oxf Engl 1993 2020. [DOI: 10.1016/j.radphyschem.2020.108881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
19
|
Tamanoi F, Matsumoto K, Doan TLH, Shiro A, Saitoh H. Studies on the Exposure of Gadolinium Containing Nanoparticles with Monochromatic X-rays Drive Advances in Radiation Therapy. NANOMATERIALS 2020; 10:nano10071341. [PMID: 32660093 PMCID: PMC7408070 DOI: 10.3390/nano10071341] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 02/07/2023]
Abstract
While conventional radiation therapy uses white X-rays that consist of a mixture of X-ray waves with various energy levels, a monochromatic X-ray (monoenergetic X-ray) has a single energy level. Irradiation of high-Z elements such as gold, silver or gadolinium with a synchrotron-generated monochromatic X-rays with the energy at or higher than their K-edge energy causes a photoelectric effect that includes release of the Auger electrons that induce DNA damage—leading to cell killing. Delivery of high-Z elements into cancer cells and tumor mass can be facilitated by the use of nanoparticles. Various types of nanoparticles containing high-Z elements have been developed. A recent addition to this growing list of nanoparticles is mesoporous silica-based nanoparticles (MSNs) containing gadolinium (Gd–MSN). The ability of Gd–MSN to inhibit tumor growth was demonstrated by evaluating effects of irradiating tumor spheroids with a precisely tuned monochromatic X-ray.
Collapse
Affiliation(s)
- Fuyuhiko Tamanoi
- Institute for Integrated Cell-Material Sciences, Institute for Advanced Study, Kyoto University, Kyoto 606-8501, Japan;
- Department of Microbio., Immunol. & Molec. Genet., University of California, Los Angeles, CA 90095, USA
- Correspondence: ; Tel.: +81-75-753-9856
| | - Kotaro Matsumoto
- Institute for Integrated Cell-Material Sciences, Institute for Advanced Study, Kyoto University, Kyoto 606-8501, Japan;
| | - Tan Le Hoang Doan
- Center for Innovative Materials and Architectures (INOMAR), Vietnam National University-Ho Chi Minh City, Ho Chi Minh City 721337, Vietnam;
| | - Ayumi Shiro
- Kansai Photon Science Institute, Quantum Beam Science Research Directorate, National Institutes for Quantum and Radiological Science and Technology, Hyogo 679-0198, Japan; (A.S.); (H.S.)
| | - Hiroyuki Saitoh
- Kansai Photon Science Institute, Quantum Beam Science Research Directorate, National Institutes for Quantum and Radiological Science and Technology, Hyogo 679-0198, Japan; (A.S.); (H.S.)
| |
Collapse
|
20
|
Maschmeyer RT, Gholami YH, Kuncic Z. Clustering effects in nanoparticle-enhanced β − emitting internal radionuclide therapy: a Monte Carlo study. Phys Med Biol 2020; 65:125007. [DOI: 10.1088/1361-6560/ab8079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
21
|
Howard D, Sebastian S, Le QVC, Thierry B, Kempson I. Chemical Mechanisms of Nanoparticle Radiosensitization and Radioprotection: A Review of Structure-Function Relationships Influencing Reactive Oxygen Species. Int J Mol Sci 2020; 21:E579. [PMID: 31963205 PMCID: PMC7013516 DOI: 10.3390/ijms21020579] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 01/19/2023] Open
Abstract
Metal nanoparticles are of increasing interest with respect to radiosensitization. The physical mechanisms of dose enhancement from X-rays interacting with nanoparticles has been well described theoretically, however have been insufficient in adequately explaining radiobiological response. Further confounding experimental observations is examples of radioprotection. Consequently, other mechanisms have gained increasing attention, especially via enhanced production of reactive oxygen species (ROS) leading to chemical-based mechanisms. Despite the large number of variables differing between published studies, a consensus identifies ROS-related mechanisms as being of significant importance. Understanding the structure-function relationship in enhancing ROS generation will guide optimization of metal nanoparticle radiosensitisers with respect to maximizing oxidative damage to cancer cells. This review highlights the physico-chemical mechanisms involved in enhancing ROS, commonly used assays and experimental considerations, variables involved in enhancing ROS generation and damage to cells and identifies current gaps in the literature that deserve attention. ROS generation and the radiobiological effects are shown to be highly complex with respect to nanoparticle physico-chemical properties and their fate within cells. There are a number of potential biological targets impacted by enhancing, or scavenging, ROS which add significant complexity to directly linking specific nanoparticle properties to a macroscale radiobiological result.
Collapse
Affiliation(s)
| | | | | | | | - Ivan Kempson
- Future Industries Institute, University of South Australia, Mawson Lakes 5095, Australia; (D.H.); (B.T.)
| |
Collapse
|
22
|
Akhtar MJ, Ahamed M, Alhadlaq H, Alrokayan S. Toxicity Mechanism of Gadolinium Oxide Nanoparticles and Gadolinium Ions in Human Breast Cancer Cells. Curr Drug Metab 2019; 20:907-917. [DOI: 10.2174/1389200220666191105113754] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 10/03/2019] [Accepted: 10/16/2019] [Indexed: 12/19/2022]
Abstract
Background:
Due to the potential advantages of Gadolinium Nanoparticles (NPs) over gadolinium elements,
gadolinium based NPs are currently being explored in the field of MRI. Either in elemental form or nanoparticulate
form, gadolinium toxicity is believed to occur due to the deposition of gadolinium ion (designated as Gd3+ ion
or simply G ion).
Objective:
There is a serious lack of literature on the mechanisms of toxicity caused by either gadolinium-based NPs
or ions. Breast cancer tumors are often subjected to MRIs, therefore, human breast cancer (MCF-7) cells could serve
as an appropriate in vitro model for the study of Gadolinium Oxide (GO) NP and G ion.
Methods:
Cytotoxicity and oxidative damage was determined by quantifying cell viability, cell membrane damage,
and Reactive Oxygen Species (ROS). Intracellular Glutathione (GSH) was measured along with cellular Total Antioxidant
Capacity (TAC). Autophagy was determined by using Monodansylcadaverine (MDC) and Lysotracker Red
(LTR) dyes in tandem. Mitochondrial Membrane Potential (MMP) was measured by JC-1 fluorescence. Physicochemical
properties of GO NPs were characterized by field emission transmission electron microscopy, X-ray diffraction,
and energy dispersive spectrum.
Results:
A time- and concentration-dependent toxicity and oxidative damage was observed due to GO NPs and G
ions. Bax/Bcl2 ratios, FITC-7AAD double staining, and cell membrane blebbing in phase-contrast images all suggested
different modes of cell death induced by NPs and ions.
Conclusion:
In summary, cell death induced by GO NPs with high aspect ratio favored apoptosis-independent cell
death, whereas G ions favored apoptosis-dependent cell death.
Collapse
Affiliation(s)
- Mohd Javed Akhtar
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh, Saudi Arabia
| | - Maqusood Ahamed
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh, Saudi Arabia
| | - Hisham Alhadlaq
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh, Saudi Arabia
| | - Salman Alrokayan
- Department of Biochemistry, College of Sciences, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
23
|
Li F, Li Z, Jin X, Liu Y, Zhang P, Li P, Shen Z, Wu A, Chen W, Li Q. Ultra-small gadolinium oxide nanocrystal sensitization of non-small-cell lung cancer cells toward X-ray irradiation by promoting cytostatic autophagy. Int J Nanomedicine 2019; 14:2415-2431. [PMID: 31040665 PMCID: PMC6455003 DOI: 10.2147/ijn.s193676] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Gadolinium-based nanoparticles (GdNPs) have been used as theranostic sensitizers in clinical radiotherapy studies; however, the biomechanisms underlying the radio-sensitizing effects of GdNPs have yet to be determined. In this study, ultra-small gadolinium oxide nanocrystals (GONs) were employed to investigate their radiosensitizing effects and biological mechanisms in non-small-cell lung cancer (NSCLC) cells under X-ray irradiation. METHOD AND MATERIALS GONs were synthesized using polyol method. Hydroxyl radical production, oxidative stress, and clonogenic survival after X-ray irradiation were used to evaluate the radiosensitizing effects of GONs. DNA double-strand breakage, cell cycle phase, and apoptosis and autophagy incidences were investigated in vitro to determine the radiosensitizing biomechanism of GONs under X-ray irradiation. RESULTS GONs induced hydroxyl radical production and oxidative stress in a dose- and concentration-dependent manner in NSCLC cells after X-ray irradiation. The sensitizer enhancement ratios of GONs ranged between 19.3% and 26.3% for the NSCLC cells under investigation with a 10% survival rate compared with that of the cells treated with irradiation alone. Addition of 3-methyladenine to the cell medium decreased the incidence rate of autophagy and increased cell survival, supporting the idea that the GONs promoted cytostatic autophagy in NSCLC cells under X-ray irradiation. CONCLUSION This study examined the biological mechanisms underlying the radiosensitizing effects of GONs on NSCLC cells and presented the first evidence for the radiosensitizing effects of GONs via activation of cytostatic autophagy pathway following X-ray irradiation.
Collapse
Affiliation(s)
- Feifei Li
- Institute of Modern Physics, Chinese Academy of Sciences, Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China, ;
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zihou Li
- University of Chinese Academy of Sciences, Beijing 100049, China
- Key Laboratory of Magnetic Materials and Devices, Chinese Academy of Sciences, Division of Functional Materials and Nano Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, Zhejiang 315201, China
| | - Xiaodong Jin
- Institute of Modern Physics, Chinese Academy of Sciences, Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China, ;
| | - Yan Liu
- Institute of Modern Physics, Chinese Academy of Sciences, Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China, ;
| | - Pengcheng Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China, ;
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ping Li
- Institute of Modern Physics, Chinese Academy of Sciences, Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China, ;
| | - Zheyu Shen
- Key Laboratory of Magnetic Materials and Devices, Chinese Academy of Sciences, Division of Functional Materials and Nano Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, Zhejiang 315201, China
| | - Aiguo Wu
- Key Laboratory of Magnetic Materials and Devices, Chinese Academy of Sciences, Division of Functional Materials and Nano Devices, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, Zhejiang 315201, China
| | - Weiqiang Chen
- Institute of Modern Physics, Chinese Academy of Sciences, Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China, ;
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences, Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China, ;
| |
Collapse
|
24
|
Abdul Rashid R, Zainal Abidin S, Khairil Anuar MA, Tominaga T, Akasaka H, Sasaki R, Kie K, Abdul Razak K, Pham BT, Hawkett BS, Carmichael MA, Geso M, Rahman WN. Radiosensitization effects and ROS generation by high Z metallic nanoparticles on human colon carcinoma cell (HCT116) irradiated under 150 MeV proton beam. OPENNANO 2019. [DOI: 10.1016/j.onano.2018.100027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
25
|
Jeon JK, Kim JK. Track analysis of a synchrotron X-ray photoelectric nanoradiator by in situ fluorescence imaging of reactive oxygen species: comparative study of gold and iron oxide nanoparticles. JOURNAL OF SYNCHROTRON RADIATION 2018; 25:1768-1773. [PMID: 30407188 DOI: 10.1107/s1600577518011396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 08/09/2018] [Indexed: 06/08/2023]
Abstract
The emission of fluorescent X-rays and low-energy electrons by mid-/high-Z nanoparticles upon irradiation with either X-ray photons or high-energy ion beams is referred to as the nanoradiator effect (NRE). A track analysis of NRE was performed using reactive oxygen species (ROS) gels, to which macrophages containing gold nanoparticles (AuNPs) were attached, together with single-cell irradiation of the intracellular nanoparticles from a microbeam of synchrotron X-rays, and the range and distribution of ^\bulletOH and O2^{ \bullet - } produced were compared with those of the Fe-nanoradiator by magnetite nanoparticles (FeONP, Fe3O4). The Au-nanoradiator generated ROS fluorescence to a greater depth and wider angle with respect to the incident X-rays than that of the Fe-nanoradiator. The ROS-oxidant fluorescence intensity ratios of ^\bulletOH to O2^{ \bullet - } were different for the AuNPs and FeONPs, reflecting different relative yields of electrons and fluorescent X-rays from NRE. In the region immediately (<100 µm) below the irradiated cell, ^\bulletOH-radicals were distributed mainly along two or three tracks in the depth direction in the FeONP- or AuNP-ROS gel. In contrast, O2^{ \bullet - } was scattered more abundantly in random directions in the AuNP-ROS gel than in the FeONP-ROS gel. Track analysis of X-ray photoelectric nanoradiator radiation showed a different range of dose distribution and relative emission compositions between Au- and Fe-nanoradiators, suggesting more extensive damage beyond a single cell containing AuNPs than one containing FeONPs.
Collapse
Affiliation(s)
- Jae Kun Jeon
- Department of Biomedical Engineering, School of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Jong Ki Kim
- Department of Biomedical Engineering, School of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| |
Collapse
|
26
|
Liu Y, Zhang P, Li F, Jin X, Li J, Chen W, Li Q. Metal-based NanoEnhancers for Future Radiotherapy: Radiosensitizing and Synergistic Effects on Tumor Cells. Theranostics 2018; 8:1824-1849. [PMID: 29556359 PMCID: PMC5858503 DOI: 10.7150/thno.22172] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 01/05/2018] [Indexed: 12/13/2022] Open
Abstract
Radiotherapy is one of the major therapeutic strategies for cancer treatment. In the past decade, there has been growing interest in using high Z (atomic number) elements (materials) as radiosensitizers. New strategies in nanomedicine could help to improve cancer diagnosis and therapy at cellular and molecular levels. Metal-based nanoparticles usually exhibit chemical inertness in cellular and subcellular systems and may play a role in radiosensitization and synergistic cell-killing effects for radiation therapy. This review summarizes the efficacy of metal-based NanoEnhancers against cancers in both in vitro and in vivo systems for a range of ionizing radiations including gamma-rays, X-rays, and charged particles. The potential of translating preclinical studies on metal-based nanoparticles-enhanced radiation therapy into clinical practice is also discussed using examples of several metal-based NanoEnhancers (such as CYT-6091, AGuIX, and NBTXR3). Also, a few general examples of theranostic multimetallic nanocomposites are presented, and the related biological mechanisms are discussed.
Collapse
Affiliation(s)
- Yan Liu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Pengcheng Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Feifei Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaodong Jin
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China
| | - Jin Li
- State Key Laboratory of Grassland Agro-ecosystems, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Weiqiang Chen
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou, China
- Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou, China
| |
Collapse
|
27
|
Seo SJ, Jeon JK, Han SM, Kim JK. Reactive oxygen species-based measurement of the dependence of the Coulomb nanoradiator effect on proton energy and atomic Z value. Int J Radiat Biol 2017; 93:1239-1247. [PMID: 28752783 DOI: 10.1080/09553002.2017.1361556] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE The Coulomb nanoradiator (CNR) effect produces the dose enhancement effects from high-Z nanoparticles under irradiation with a high-energy ion beam. To gain insight into the radiation dose and biological significance of the CNR effect, the enhancement of reactive oxygen species (ROS) production from iron oxide or gold NPs (IONs or AuNPs, respectively) in water was investigated using traversing proton beams. METHODS AND MATERIALS The dependence of nanoradiator-enhanced ROS production on the atomic Z value and proton energy was investigated. Two biologically important ROS species were measured using fluorescent probes specific to •OH or [Formula: see text] in a series of water phantoms containing either AuNPs or IONs under irradiation with a 45- or 100-MeV proton beam. RESULTS The enhanced generation of hydroxyl radicals (•OH) and superoxide anions ([Formula: see text]) was determined to be caused by the dependence on the NP concentration and proton energy. The proton-induced Au or iron oxide nanoradiators exhibited different ROS enhancement rates depending on the proton energy, suggesting that the CNR radiation varied. The curve of the superoxide anion production from the Au-nanoradiator showed strong non-linearity, unlike the linear behavior observed for hydroxyl radical production and the X-ray photoelectric nanoradiator. In addition, the 45-MeV proton-induced Au nanoradiator exhibited an ROS enhancement ratio of 8.54/1.50 ([Formula: see text] / •OH), similar to that of the 100-KeV X-ray photoelectric Au nanoradiator (7.68/1.46). CONCLUSIONS The ROS-based detection of the CNR effect revealed its dependence on the proton beam energy, dose and atomic Z value and provided insight into the low-linear energy transfer (LET) CNR radiation, suggesting that these factors may influence the therapeutic efficacy via chemical reactivities, transport behaviors, and intracellular oxidative stress.
Collapse
Affiliation(s)
- Seung-Jun Seo
- a Department of Biomedical Engineering and Radiology, School of Medicine , Catholic University of Daegu , Daegu City , South Korea
| | - Jae-Kun Jeon
- a Department of Biomedical Engineering and Radiology, School of Medicine , Catholic University of Daegu , Daegu City , South Korea
| | - Sung-Mi Han
- b Department of Anatomy, School of Medicine , Catholic University of Daegu , Daegu City , South Korea
| | - Jong-Ki Kim
- a Department of Biomedical Engineering and Radiology, School of Medicine , Catholic University of Daegu , Daegu City , South Korea
| |
Collapse
|
28
|
Wozny AS, Aloy MT, Alphonse G, Magné N, Janier M, Tillement O, Lux F, Beuve M, Rodriguez-Lafrasse C. Gadolinium-based nanoparticles as sensitizing agents to carbon ions in head and neck tumor cells. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:2655-2660. [PMID: 28779947 DOI: 10.1016/j.nano.2017.07.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 06/13/2017] [Accepted: 07/24/2017] [Indexed: 12/31/2022]
Abstract
Hadrontherapy presents the major advantage of improving tumor sterilization while sparing surrounding healthy tissues because of the particular ballistic (Bragg peak) of carbon ions. However, its efficacy is still limited in the most resistant cancers, such as grade III-IV head and neck squamous cell carcinoma (HNSCC), in which the association of carbon ions with gadolinium-based nanoparticles (AGuIX®) could be used as a Trojan horse. We report for the first time the radioenhancing effect of AGuIX® when combined with carbon ion irradiation in human tumor cells. An increase in relative biological effectiveness (1.7) in three HNSCC cell lines (SQ20B, FaDu, and Cal33) was associated with a significant reduction in the radiation dose needed for killing cells. Radiosensitization goes through a higher number of unrepaired DNA double-strand breaks. These results underline the strong potential of AGuIX® in sensitizing aggressive tumors to hadrontherapy and, therefore, improving local control while lowering acute/late toxicity.
Collapse
Affiliation(s)
- Anne-Sophie Wozny
- Univ Lyon, Université Lyon 1, UMR CNRS5822/IN2P3, IPNL, PRISME, Radiobiologie Cellulaire et Moléculaire, Faculté de Médecine Lyon-Sud, Oullins cedex, France; Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre Bénite, France
| | - Marie-Thérèse Aloy
- Univ Lyon, Université Lyon 1, UMR CNRS5822/IN2P3, IPNL, PRISME, Radiobiologie Cellulaire et Moléculaire, Faculté de Médecine Lyon-Sud, Oullins cedex, France
| | - Gersende Alphonse
- Univ Lyon, Université Lyon 1, UMR CNRS5822/IN2P3, IPNL, PRISME, Radiobiologie Cellulaire et Moléculaire, Faculté de Médecine Lyon-Sud, Oullins cedex, France; Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre Bénite, France
| | - Nicolas Magné
- Univ Lyon, Université Lyon 1, UMR CNRS5822/IN2P3, IPNL, PRISME, Radiobiologie Cellulaire et Moléculaire, Faculté de Médecine Lyon-Sud, Oullins cedex, France; Département de Radiothérapie, Institut de Cancérologie de la Loire Lucien Neuwirth, St Priest en Jarez, France
| | - Marc Janier
- Univ Lyon, Université Lyon1, CNRS, LAGEP UMR 5007, IMTHERNAT, Hôpital Edouard Herriot, Lyon, France
| | - Olivier Tillement
- Univ Lyon, Université Lyon 1, Institut Lumière Matière, UMR 5306 CNRS, Villeurbanne cedex, France
| | - François Lux
- Univ Lyon, Université Lyon 1, Institut Lumière Matière, UMR 5306 CNRS, Villeurbanne cedex, France
| | - Michael Beuve
- Univ Lyon, Université Lyon 1, UMR CNRS5822/IN2P3, IPNL, PRISME, PHABIO, Villeurbanne, France
| | - Claire Rodriguez-Lafrasse
- Univ Lyon, Université Lyon 1, UMR CNRS5822/IN2P3, IPNL, PRISME, Radiobiologie Cellulaire et Moléculaire, Faculté de Médecine Lyon-Sud, Oullins cedex, France; Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre Bénite, France.
| |
Collapse
|
29
|
Jeon JK, Han SM, Min SK, Seo SJ, Ihm K, Chang WS, Kim JK. Coulomb nanoradiator-mediated, site-specific thrombolytic proton treatment with a traversing pristine Bragg peak. Sci Rep 2016; 6:37848. [PMID: 27897205 PMCID: PMC5126678 DOI: 10.1038/srep37848] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 10/31/2016] [Indexed: 12/28/2022] Open
Abstract
Traversing proton beam-irradiated, mid/high-Z nanoparticles produce site-specific enhancement of X-ray photon-electron emission via the Coulomb nanoradiator (CNR) effect, resulting in a nano- to micro-scale therapeutic effect at the nanoparticle-uptake target site. Here, we demonstrate the uptake of iron oxide nanoparticles (IONs) and nanoradiator-mediated, site-specific thrombolysis without damaging the vascular endothelium in an arterial thrombosis mouse model. The enhancement of low-energy electron (LEE) emission and reactive oxygen species (ROS) production from traversing proton beam-irradiated IONs was examined. Flow recovery was only observed in CNR-treated mice, and greater than 50% removal of the thrombus was achieved. A 2.5-fold greater reduction in the thrombus-enabled flow recovery was observed in the CNR group compared with that observed in the untreated ION-only and proton-only control groups (p < 0.01). Enhancement of the X-ray photon-electron emission was evident from both the pronounced Shirley background in the electron yield and the 1.2- to 2.5-fold enhanced production of ROS by the proton-irradiated IONs, which suggests chemical degradation of the thrombus without potent emboli.
Collapse
Affiliation(s)
- Jae-Kun Jeon
- Departments of Biomedical Engineering, Catholic University of Daegu, School of Medicine, Daegu, Korea
| | - Sung-Mi Han
- Anatomy, and Diagnostic Imaging, Catholic University of Daegu, School of Medicine, Daegu, Korea
| | - Soon-Ki Min
- Catholic University of Daegu, School of Medicine, Daegu, Korea
| | - Seung-Jun Seo
- Departments of Biomedical Engineering, Catholic University of Daegu, School of Medicine, Daegu, Korea
| | - Kyuwook Ihm
- Pohang Accelerator Laboratory, Pohang, Korea
| | - Won-Seok Chang
- Departments of Biomedical Engineering, Catholic University of Daegu, School of Medicine, Daegu, Korea
| | - Jong-Ki Kim
- Departments of Biomedical Engineering, Catholic University of Daegu, School of Medicine, Daegu, Korea
| |
Collapse
|
30
|
Key clinical beam parameters for nanoparticle-mediated radiation dose amplification. Sci Rep 2016; 6:34040. [PMID: 27658637 PMCID: PMC5034311 DOI: 10.1038/srep34040] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 09/07/2016] [Indexed: 11/12/2022] Open
Abstract
As nanoparticle solutions move towards human clinical trials in radiation therapy, the influence of key clinical beam parameters on therapeutic efficacy must be considered. In this study, we have investigated the clinical radiation therapy delivery variables that may significantly affect nanoparticle-mediated radiation dose amplification. We found a benefit for situations which increased the proportion of low energy photons in the incident beam. Most notably, “unflattened” photon beams from a clinical linear accelerator results in improved outcomes relative to conventional “flat” beams. This is measured by significant DNA damage, tumor growth suppression, and overall improvement in survival in a pancreatic tumor model. These results, obtained in a clinical setting, clearly demonstrate the influence and importance of radiation therapy parameters that will impact clinical radiation dose amplification with nanoparticles.
Collapse
|
31
|
Jeon JK, Han SM, Kim JK. Fluorescence imaging of reactive oxygen species by confocal laser scanning microscopy for track analysis of synchrotron X-ray photoelectric nanoradiator dose: X-ray pump-optical probe. JOURNAL OF SYNCHROTRON RADIATION 2016; 23:1191-1196. [PMID: 27577774 DOI: 10.1107/s1600577516009103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 06/06/2016] [Indexed: 06/06/2023]
Abstract
Bursts of emissions of low-energy electrons, including interatomic Coulomb decay electrons and Auger electrons (0-1000 eV), as well as X-ray fluorescence produced by irradiation of large-Z element nanoparticles by either X-ray photons or high-energy ion beams, is referred to as the nanoradiator effect. In therapeutic applications, this effect can damage pathological tissues that selectively take up the nanoparticles. Herein, a new nanoradiator dosimetry method is presented that uses probes for reactive oxygen species (ROS) incorporated into three-dimensional gels, on which macrophages containing iron oxide nanoparticles (IONs) are attached. This method, together with site-specific irradiation of the intracellular nanoparticles from a microbeam of polychromatic synchrotron X-rays (5-14 keV), measures the range and distribution of OH radicals produced by X-ray emission or superoxide anions ({\rm{O}}_2^-) produced by low-energy electrons. The measurements are based on confocal laser scanning of the fluorescence of the hydroxyl radical probe 2-[6-(4'-amino)phenoxy-3H-xanthen-3-on-9-yl] benzoic acid (APF) or the superoxide probe hydroethidine-dihydroethidium (DHE) that was oxidized by each ROS, enabling tracking of the radiation dose emitted by the nanoradiator. In the range 70 µm below the irradiated cell, ^\bullet{\rm{OH}} radicals derived mostly from either incident X-ray or X-ray fluorescence of ION nanoradiators are distributed along the line of depth direction in ROS gel. In contrast, {\rm{O}}_2^- derived from secondary electron or low-energy electron emission by ION nanoradiators are scattered over the ROS gel. ROS fluorescence due to the ION nanoradiators was observed continuously to a depth of 1.5 mm for both oxidized APF and oxidized DHE with relatively large intensity compared with the fluorescence caused by the ROS produced solely by incident primary X-rays, which was limited to a depth of 600 µm, suggesting dose enhancement as well as more penetration by nanoradiators. In conclusion, the combined use of a synchrotron X-ray microbeam-irradiated three-dimensional ROS gel and confocal laser scanning fluorescence microscopy provides a simple dosimetry method for track analysis of X-ray photoelectric nanoradiator radiation, suggesting extensive cellular damage with dose-enhancement beyond a single cell containing IONs.
Collapse
Affiliation(s)
- Jae Kun Jeon
- Department of Biomedical Engineering, School of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Sung Mi Han
- Department of Anatomy, School of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Jong Ki Kim
- Department of Biomedical Engineering, School of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| |
Collapse
|