1
|
Zhang X, Qian X, Zhao Y, Ye M, Li L, Chu J. Identification and analysis of prognostic ion homeostasis characteristics in kidney renal clear cell carcinoma. Heliyon 2025; 11:e41736. [PMID: 39897849 PMCID: PMC11782977 DOI: 10.1016/j.heliyon.2025.e41736] [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: 07/15/2024] [Revised: 01/04/2025] [Accepted: 01/05/2025] [Indexed: 02/04/2025] Open
Abstract
Kidney renal clear cell carcinoma (KIRC), a prevalent primary malignant tumor within the urinary system, is characterized by significant heterogeneity. It has been shown that ion channels are important targets for anti-tumor therapy. In this study, we screened 70 selected KIRC related ion homeostasis genes with significant differential expression. We established diagnostic and prognostic models for 15 genes (PDK4, JPH4, ATP1A3, CCL7, CYP27B1, ABCB6, TNFSF11, MCHR1, TNNI3, ANGPTL3, Ednrb, SAA1, Chrna9, TMPRSS6, CCL14) by LASSO regression in the TCGA-KIRC cohort. We also provided a nomogram based on ion homeostasis for clinicians to explore the combined effect of the risk model on clinical variables. Patients in the low-risk group have a significant survival advantage. The potential clinical benefit of our predicted 15 gene signatures in clinical strategies was validated by Calibration Curves and DCA curves. Ultimately, the immune microenvironment and enrichment pathways were analyzed among individuals categorized as high-risk and low-risk. The predictable ion homeostasis-associated 15 gene signature established in this study predicts overall survival outcomes in patients with KIRC, to some extent helping clinicians to select personalized treatment regimens.
Collapse
Affiliation(s)
| | | | - Yong Zhao
- Department of Urology, Shanghai Baoshan Luodian Hospital, Baoshan District, Shanghai, 201908, China
| | - Maofei Ye
- Department of Urology, Shanghai Baoshan Luodian Hospital, Baoshan District, Shanghai, 201908, China
| | - Liyang Li
- Department of Urology, Shanghai Baoshan Luodian Hospital, Baoshan District, Shanghai, 201908, China
| | - Jian Chu
- Department of Urology, Shanghai Baoshan Luodian Hospital, Baoshan District, Shanghai, 201908, China
| |
Collapse
|
2
|
Zhou H, Yang P, Zhang T, Kepp O, Ren Y, Jiang N, Liu R, Li J, Li C. The role of apoptosis, immunogenic cell death, and macrophage polarization in carbon ion radiotherapy for keloids: Targeting the TGF-β1/SMADs signaling pathway. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167499. [PMID: 39245184 DOI: 10.1016/j.bbadis.2024.167499] [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: 05/04/2024] [Revised: 08/11/2024] [Accepted: 08/23/2024] [Indexed: 09/10/2024]
Abstract
Keloids, characterized by excessive extracellular matrix (ECM) deposition and aberrant fibrous tissue proliferation, present significant therapeutic challenges due to their recalcitrant and recurrent nature. This study explores the efficacy of Carbon Ion Radiotherapy (CIRT) as a novel therapeutic approach for keloids, focusing on its impact on fibroblast proliferation, apoptosis induction, immunogenic cell death (ICD), macrophage polarization, and the TGF-β/SMAD signaling pathway. Utilizing a murine model of keloid formed by subcutaneous injection of zeocin in C57BL/6 mice, we demonstrated that CIRT effectively reduces collagenous fiber synthesis and collagen production in keloid tissues. Further, CIRT was shown to inhibit keloid fibroblast proliferation and to induce apoptosis, as evidenced by increased expression of apoptosis-related proteins and confirmed through flow cytometry and TUNEL assay. Notably, CIRT induced mitochondrial stress, leading to enhanced immunogenicity of cell death, characterized by increased expression of ICD markers and secretion of interferon-γ. Additionally, CIRT promoted a shift from M2 to M1 macrophage polarization, potentially reducing TGF-β release and mitigating ECM deposition. Our findings suggest that CIRT mediates its therapeutic effects through the inhibition of the TGF-β/SMAD signaling pathway, thereby attenuating ECM formation and offering a promising avenue for keloid treatment. This study underscores the potential of CIRT as an innovative strategy for managing keloids, highlighting its multifaceted impact on key cellular processes involved in keloid pathogenesis.
Collapse
Affiliation(s)
- Heng Zhou
- School of Public Health, Yangzhou University, Yangzhou, China; Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Pengfei Yang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Tianyi Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Oliver Kepp
- Centre de Recherche des Cordeliers, Université de Paris, INSERM U1138, Paris, France
| | - Yanxian Ren
- School of Public Health, Yangzhou University, Yangzhou, China; The First Hospital of Lanzhou University, Lanzhou, China
| | - Ningzu Jiang
- School of Public Health, Yangzhou University, Yangzhou, China; The First Hospital of Lanzhou University, Lanzhou, China
| | - Ruifeng Liu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China; Renmin Hospital of Wuhan Economic and Technological Development Zone, Wuhan, China
| | - Jin Li
- Renmin Hospital of Wuhan Economic and Technological Development Zone, Wuhan, China
| | - Chenghao Li
- School of Public Health, Yangzhou University, Yangzhou, China; Yangzhou University Medical College, Yangzhou, China..
| |
Collapse
|
3
|
Yoshimoto M, Washiyama K, Ohnuki K, Doi A, Inokuchi M, Kojima M, Miller BW, Yoshii Y, Inaki A, Fujii H. Long-Term Therapeutic Effects of 225Ac-DOTA-E[c(RGDfK)] 2 Induced by Radiosensitization via G2/M Arrest in Pancreatic Ductal Adenocarcinoma. Pharmaceutics 2024; 17:9. [PMID: 39861661 PMCID: PMC11768328 DOI: 10.3390/pharmaceutics17010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 01/27/2025] Open
Abstract
Background: Alpha radionuclide therapy has emerged as a promising novel strategy for cancer treatment; however, the therapeutic potential of 225Ac-labeled peptides in pancreatic cancer remains uninvestigated. Methods: In the cytotoxicity study, tumor cells were incubated with 225Ac-DOTA-RGD2. DNA damage responses (γH2AX and 53BP1) were detected using flowcytometry or immunohistochemistry analysis. Biodistribution and therapeutic studies were carried out in BxPC-3-bearing mice. Results: 225Ac-DOTA-RGD2 demonstrated potent cytotoxicity against cells expressing αvβ3 or αvβ6 integrins and induced G2/M arrest and γH2AX expression as a marker of double-stranded DNA breaks. 225Ac-DOTA-RGD2 (20, 40, 65, or 90 kBq) showed favorable pharmacokinetics and remarkable tumor growth inhibition without severe side effects in the BxPC-3 mouse model. In vitro studies revealed that 5 and 10 kBq/mL of 225Ac-DOTA-RGD2 swiftly induced G2/M arrest and elevated γH2AX expression. Furthermore, to clarify the mechanism of successful tumor growth inhibition for a long duration in vivo, we investigated whether short-term high radiation exposure enhances radiation sensitivity. Initially, a 4 h induction treatment with 5 and 10 kBq/mL of 225Ac-DOTA-RGD2 enhanced both cytotoxicity and γH2AX expression with 0.5 kBq/mL of 225Ac-DOTA-RGD2 compared to a treatment with only 0.5 kBq/mL of 225Ac-DOTA-RGD2. Meanwhile, the γH2AX expression induced by 5 or 10 kBq/mL of 225Ac-DOTA-RGD2 alone decreased over time. Conclusions: These findings highlight the potential of using 225Ac-DOTA-RGD2 in the treatment of intractable pancreatic cancers, as its ability to induce G2/M cell cycle arrest enhances radiosensitization, resulting in notable growth inhibition.
Collapse
Affiliation(s)
- Mitsuyoshi Yoshimoto
- Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Kohshin Washiyama
- Advanced Clinical Research Center, Fukushima Global Medical Science Center, Fukushima Medical University, Fukushima 960-1295, Japan;
| | - Kazunobu Ohnuki
- Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Ayano Doi
- Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Miki Inokuchi
- Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Motohiro Kojima
- Division of Pathology, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Brian W. Miller
- Department of Radiation Oncology, College of Medicine, University of Arizona, Tucson, AZ 85719, USA
| | - Yukie Yoshii
- National Institute of Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Anri Inaki
- Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| | - Hirofumi Fujii
- Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa 277-8577, Japan
| |
Collapse
|
4
|
Yu B, Li KW, Fan Y, Pei X. Value of Carbon-Ion Radiation Therapy for Breast Cancer. Int J Part Ther 2024; 14:100629. [PMID: 39309411 PMCID: PMC11415881 DOI: 10.1016/j.ijpt.2024.100629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/19/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
To explore the challenges and future of carbon-ion radiation therapy (CIRT) in breast cancer, we summarized the progress of nonclinical and clinical studies on CIRT for breast cancer in this review. A total of 6 nonclinical studies have been reported, which demonstrated a better effect of Carbon-ion irradiation compared with X-ray in breast cancer cell lines (including triple-negative breast cancer and Human Epidermal Growth Factor Receptor 2-negative breast cancer). Combination with Hh inhibitor, dual tyrosine kinase inhibitor, and PARP inhibitor is promising as demonstrated in the in vitro studies. Approximately 34 patients with breast cancer went through CIRT treatment, as reported in 5 clinical studies. All studies demonstrated promising treatment effects with acceptable and manageable risks. In these studies, a total of 21 patients were reported with post-treatment response assessments, among whom 19 patients (90.48%) reported a response of complete response or partial response. The complete response rate was 66.67%. The time to complete the response ranged from 3 months to 24 months. No adverse events were observed in these studies except for grade 1 acute skin reaction in 14 out of the 21 patients (66.67%). Although the time to respond was longer than expected in some studies, the persistent responses and satisfactory safety profile provided the rationale for further research on this new therapy.
Collapse
Affiliation(s)
- Bowen Yu
- Galactophore Department, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Kai-Wen Li
- Department of Technology, CAS Ion Medical Technology Co., Ltd., Beijing, China
| | - Yingyi Fan
- Galactophore Department, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Xiaohua Pei
- Beijing University of Chinese Medicine Xiamen Hospital, Xiamen, China
| |
Collapse
|
5
|
Musielak M, Graczyk K, Liszka M, Christou A, Rosochowicz MA, Lach MS, Adamczyk B, Suchorska WM, Piotrowski T, Stenerlöw B, Malicki J. Impact of Proton Irradiation Depending on Breast Cancer Subtype in Patient-Derived Cell Lines. Int J Mol Sci 2024; 25:10494. [PMID: 39408826 PMCID: PMC11477436 DOI: 10.3390/ijms251910494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Research on different types of ionizing radiation's effects has been ongoing for years, revealing its efficacy in damaging cancer cells. Solid tumors comprise diverse cell types, each being able to respond differently to radiation. This study evaluated the radiobiological response of established (MDA-MB-231 (Triple negative breast cancer, TNBC), MCF-7 (Luminal A)) and patient-derived malignant cell lines, cancer-associated fibroblasts, and skin fibroblasts following proton IRR. All cell line types were irradiated with the proton dose of 2, 4, and 6 Gy. The radiobiological response was assessed using clonogenic assay, γH2AX, and p53 staining. It was noticeable that breast cancer lines of different molecular subtypes displayed no significant variations in their response to proton IRR. In terms of cancer-associated fibroblasts extracted from the tumor tissue, the line derived from a TNBC subtype tumor demonstrated higher resistance to ionizing radiation compared to lines isolated from luminal A tumors. Fibroblasts extracted from patients' skin responded identically to all doses of proton radiation. This study emphasizes that tumor response is not exclusively determined by the elimination of breast cancer cells, but also takes into account tumor microenvironmental variables and skin reactions.
Collapse
Affiliation(s)
- Marika Musielak
- Department of Electroradiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (W.M.S.); (T.P.); (J.M.)
- Doctoral School, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
- Radiobiology Laboratory, Department of Medical Physics, Greater Poland Cancer Centre, 61-866 Poznan, Poland;
| | - Kinga Graczyk
- Clinical Dosimetry, Department of Medical Physics, Greater Poland Cancer Centre, 61-866 Poznan, Poland;
- The Skandion Clinic, 751 23 Uppsala, Sweden; (M.L.); (A.C.)
| | | | | | - Monika A. Rosochowicz
- Doctoral School, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
- Radiobiology Laboratory, Department of Medical Physics, Greater Poland Cancer Centre, 61-866 Poznan, Poland;
- Department of Orthopaedics and Traumatology, Poznan University of Medical Sciences, 61-701 Poznan, Poland
| | - Michał S. Lach
- Radiobiology Laboratory, Department of Medical Physics, Greater Poland Cancer Centre, 61-866 Poznan, Poland;
| | - Beata Adamczyk
- Breast Surgical Oncology Department, Greater Poland Cancer Centre, 61-866 Poznan, Poland;
| | - Wiktoria M. Suchorska
- Department of Electroradiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (W.M.S.); (T.P.); (J.M.)
- Radiobiology Laboratory, Department of Medical Physics, Greater Poland Cancer Centre, 61-866 Poznan, Poland;
| | - Tomasz Piotrowski
- Department of Electroradiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (W.M.S.); (T.P.); (J.M.)
- Department of Medical Physics, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Bo Stenerlöw
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 753 10 Uppsala, Sweden;
| | - Julian Malicki
- Department of Electroradiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland; (W.M.S.); (T.P.); (J.M.)
- Department of Medical Physics, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| |
Collapse
|
6
|
Talaee O, Faghihi R, Rastegari B, Sina S. Enhanced radio-photodynamic therapy potential of advanced gold-based nanoclusters for breast cancer treatment. Radiol Phys Technol 2024; 17:703-714. [PMID: 39014282 DOI: 10.1007/s12194-024-00824-8] [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: 02/06/2024] [Revised: 05/31/2024] [Accepted: 06/23/2024] [Indexed: 07/18/2024]
Abstract
The purpose of current study was to assess the impact of ALA-coated gold nanoclusters (Au NPs) on the combined therapeutic effects of radiotherapy (RT) and photodynamic therapy (PDT) on healthy MCF-10A and MCF-7 breast cancer cells. The Au NPs were covered with ALA using PEG polymer, resulting in the synthesis of Au@ALA NPs. The successful synthesis of the final NPs was confirmed through FTIR, XRD, TEM, and UV-Vis tests. MCF-10A and MCF-7 cell lines were treated with different concentrations of Au@ALA NPs and exposed to irradiation of 2 and 4 Gy (using MV X-ray) and 630 nm laser light irradiation. Cytotoxicity was assessed using a multifaceted approach involving the MTT assay, real-time PCR, and colony forming assay. The findings revealed that the damage inflicted by Au@ALA NPs on cancerous tissue was significantly greater than that on normal tissue. The cytotoxic effects of all experimental groups exhibited a direct correlation with increasing concentrations and radiation doses. The combination of Au@ALA NPs with RT doses of 2 and 4 Gy resulted in a reduction in cell viability by a factor of 1.58 (P = 0.001) and 1.73 (P = 0.004), respectively. Furthermore, the simultaneous intervention of NPs with PDT and RT at doses of 2 and 4 Gy led to a decrease in cell viability by a factor of 2.10 (P = 0.001) and 3.08 (P = 0.001) in turn. Furthermore, the real-time PCR and colonogenic assay results demonstrated that the combined treatment significantly increased phosphorylation of ATM and expression of TP53, indicating an adequate synergistic effect on breast cancer cells. The concurrent application of Au@ALA NPs in RT and PDT successfully enhanced the radiosensitization of breast cancer cells to megavoltage RT and PDT.
Collapse
Affiliation(s)
- Omid Talaee
- Nuclear Engineering Department, Shiraz University, Shiraz, Iran
| | - Reza Faghihi
- Nuclear Engineering Department, Shiraz University, Shiraz, Iran.
- Radiation Research Center, Shiraz University, Shiraz, Iran.
| | - Banafsheh Rastegari
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sedigheh Sina
- Nuclear Engineering Department, Shiraz University, Shiraz, Iran
- Radiation Research Center, Shiraz University, Shiraz, Iran
| |
Collapse
|
7
|
Donato L, Mordà D, Scimone C, Alibrandi S, D'Angelo R, Sidoti A. From powerhouse to regulator: The role of mitoepigenetics in mitochondrion-related cellular functions and human diseases. Free Radic Biol Med 2024; 218:105-119. [PMID: 38565400 DOI: 10.1016/j.freeradbiomed.2024.03.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/26/2024] [Accepted: 03/30/2024] [Indexed: 04/04/2024]
Abstract
Beyond their crucial role in energy production, mitochondria harbor a distinct genome subject to epigenetic regulation akin to that of nuclear DNA. This paper delves into the nascent but rapidly evolving fields of mitoepigenetics and mitoepigenomics, exploring the sophisticated regulatory mechanisms governing mitochondrial DNA (mtDNA). These mechanisms encompass mtDNA methylation, the influence of non-coding RNAs (ncRNAs), and post-translational modifications of mitochondrial proteins. Together, these epigenetic modifications meticulously coordinate mitochondrial gene transcription, replication, and metabolism, thereby calibrating mitochondrial function in response to the dynamic interplay of intracellular needs and environmental stimuli. Notably, the dysregulation of mitoepigenetic pathways is increasingly implicated in mitochondrial dysfunction and a spectrum of human pathologies, including neurodegenerative diseases, cancer, metabolic disorders, and cardiovascular conditions. This comprehensive review synthesizes the current state of knowledge, emphasizing recent breakthroughs and innovations in the field. It discusses the potential of high-resolution mitochondrial epigenome mapping, the diagnostic and prognostic utility of blood or tissue mtDNA epigenetic markers, and the promising horizon of mitochondrial epigenetic drugs. Furthermore, it explores the transformative potential of mitoepigenetics and mitoepigenomics in precision medicine. Exploiting a theragnostic approach to maintaining mitochondrial allostasis, this paper underscores the pivotal role of mitochondrial epigenetics in charting new frontiers in medical science.
Collapse
Affiliation(s)
- Luigi Donato
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, 98122, Messina, Italy; Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Euro-Mediterranean Institute of Science and Technology (I.E.ME.S.T.) 90139 Palermo, Italy.
| | - Domenico Mordà
- Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Euro-Mediterranean Institute of Science and Technology (I.E.ME.S.T.) 90139 Palermo, Italy; Department of Veterinary Sciences, University of Messina, 98122, Messina, Italy.
| | - Concetta Scimone
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, 98122, Messina, Italy; Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Euro-Mediterranean Institute of Science and Technology (I.E.ME.S.T.) 90139 Palermo, Italy.
| | - Simona Alibrandi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, 98122, Messina, Italy; Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Euro-Mediterranean Institute of Science and Technology (I.E.ME.S.T.) 90139 Palermo, Italy.
| | - Rosalia D'Angelo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, 98122, Messina, Italy.
| | - Antonina Sidoti
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Division of Medical Biotechnologies and Preventive Medicine, University of Messina, 98122, Messina, Italy.
| |
Collapse
|
8
|
Zhou Z, Guan B, Xia H, Zheng R, Xu B. Particle radiotherapy in the era of radioimmunotherapy. Cancer Lett 2023:216268. [PMID: 37331583 DOI: 10.1016/j.canlet.2023.216268] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/24/2023] [Accepted: 06/11/2023] [Indexed: 06/20/2023]
Abstract
Radiotherapy (RT) is one of the key modalities for cancer treatment, and more than 70% of tumor patients will receive RT during the course of their disease. Particle radiotherapy, such as proton radiotherapy, carbon-ion radiotherapy (CIRT) and boron neutron capture therapy (BNCT), is currently available for the treatment of patients Immunotherapy combined with photon RT has been successfully used in the clinic. The effect of immunotherapy combined with particle RT is an area of interest. However, the molecular mechanisms underlying the effects of combined immunotherapy and particle RT remain largely unknown. In this review, we summarize the properties of different types of particle RT and the mechanisms underlying their radiobiological effects. Additionally, we compared the main molecular players in photon RT and particle RT and the mechanisms involved the RT-mediated immune response.
Collapse
Affiliation(s)
- Zihan Zhou
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Xinquan Road 29, Fuzhou, 350000, Fuzhou, China.
| | - Bingjie Guan
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Xinquan Road 29, Fuzhou, 350000, Fuzhou, China.
| | - Huang Xia
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Xinquan Road 29, Fuzhou, 350000, Fuzhou, China.
| | - Rong Zheng
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Xinquan Road 29, Fuzhou, 350000, Fuzhou, China; Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Fuzhou, Xinquan Road 29, Fuzhou, 350000, Fujian, China; Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies), Fuzhou, Xinquan Road 29, Fuzhou, 350000, Fujian, China.
| | - Benhua Xu
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Xinquan Road 29, Fuzhou, 350000, Fuzhou, China; Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for Tumors (Fujian Medical University), Fuzhou, Xinquan Road 29, Fuzhou, 350000, Fujian, China; Clinical Research Center for Radiology and Radiotherapy of Fujian Province (Digestive, Hematological and Breast Malignancies), Fuzhou, Xinquan Road 29, Fuzhou, 350000, Fujian, China.
| |
Collapse
|
9
|
Manunu B, Serafin AM, Akudugu JM. BAG1, MGMT, FOXO1, and DNAJA1 as potential drug targets for radiosensitizing cancer cell lines. Int J Radiat Biol 2023; 99:292-307. [PMID: 35511481 DOI: 10.1080/09553002.2022.2074164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND AND PURPOSE Activation of some signaling pathways can promote cell survival and have a negative impact on tumor response to radiotherapy. Here, the role of differences in expression levels of genes related to the poly(ADP-ribose) polymerase-1 (PARP-1), heat shock protein 90 (Hsp90), B-cell lymphoma 2 (Bcl-2), and phosphoinositide 3-kinase (PI3K) pathways in the survival or death of cells following X-ray exposure was investigated. METHODS Eight human cell cultures (MCF-7 and MDA-MB-231: breast cancers; MCF-12A: apparently normal breast; A549: lung cancer; L132: normal lung; G28, G44 and G112: glial cancers) were irradiated with X-rays. The colony-forming and real-time PCR based on a custom human pathway RT2 Profiler PCR Array assays were used to evaluate cell survival and gene expression, respectively. RESULTS The surviving fractions at 2 Gy for the cell lines, in order of increasing radioresistance, were found to be as follows: MCF-7 (0.200 ± 0.011), G44 (0.277 ± 0.065), L132 (0.367 ± 0.023), MDA-MB-231 (0.391 ± 0.057), G112 (0.397 ± 0.113), A549 (0.490 ± 0.048), MCF-12A (0.526 ± 0.004), and G28 (0.633 ± 0.094). The rank order of radioresistance at 6 Gy was: MCF-7 < L132 < G44 < MDA-MB-231 < A549 < G28 < G112 < MCF-12A. PCR array data analysis revealed that several genes were differentially expressed between irradiated and unirradiated cell cultures. The following genes, with fold changes: BCL2A1 (21.91), TP53 (8743.75), RAD51 (11.66), FOX1 (65.86), TCP1 (141.32), DNAJB1 (3283.64), RAD51 (51.52), and HSPE1 (12887.29) were highly overexpressed, and BAX (-127.21), FOX1 (-81.79), PDPK1 (-1241.78), BRCA1 (-8.70), MLH1 (-12143.95), BCL2 (-18.69), CCND1 (-46475.98), and GJA1 (-2832.70) were highly underexpressed in the MDA-MB-231, MCF-7, MCF-12A, A549, L132, G28, G44, and G112 cell lines, respectively. The radioresistance in the malignant A549 and G28 cells was linked to upregulation in the apoptotic, DNA repair, PI3K, and Hsp90 pathway genes BAG1, MGMT, FOXO1, and DNAJA1, respectively, and inhibition of these genes resulted in significant radiosensitization. CONCLUSIONS Targeting BAG1, MGMT, FOXO1, and DNAJA1 with specific inhibitors might effectively sensitize radioresistant tumors to radiotherapy.
Collapse
Affiliation(s)
- Bayanika Manunu
- Division of Radiobiology, Department of Medical Imaging and Clinical Oncology, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
| | - Antonio M Serafin
- Division of Radiobiology, Department of Medical Imaging and Clinical Oncology, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
| | - John M Akudugu
- Division of Radiobiology, Department of Medical Imaging and Clinical Oncology, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
| |
Collapse
|
10
|
The 'stealth-bomber' paradigm for deciphering the tumour response to carbon-ion irradiation. Br J Cancer 2023; 128:1429-1438. [PMID: 36639527 PMCID: PMC10070470 DOI: 10.1038/s41416-022-02117-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/01/2022] [Accepted: 12/08/2022] [Indexed: 01/14/2023] Open
Abstract
Numerous studies have demonstrated the higher biological efficacy of carbon-ion irradiation (C-ions) and their ballistic precision compared with photons. At the nanometre scale, the reactive oxygen species (ROS) produced by radiation and responsible for the indirect effects are differentially distributed according to the type of radiation. Photon irradiation induces a homogeneous ROS distribution, whereas ROS remain condensed in clusters in the C-ions tracks. Based on this linear energy transfer-dependent differential nanometric ROS distribution, we propose that the higher biological efficacy and specificities of the molecular response to C-ions rely on a 'stealth-bomber' effect. When biological targets are on the trajectories of the particles, the clustered radicals in the tracks are responsible for a 'bomber' effect. Furthermore, the low proportion of ROS outside the tracks is not able to trigger the cellular mechanisms of defence and proliferation. The ability of C-ions to deceive the cellular defence of the cancer cells is then categorised as a 'stealth' effect. This review aims to classify the biological arguments supporting the paradigm of the 'stealth-bomber' as responsible for the biological superiority of C-ions compared with photons. It also explains how and why C-ions will always be more efficient for treating patients with radioresistant cancers than conventional radiotherapy.
Collapse
|
11
|
Helm A, Totis C, Durante M, Fournier C. Are charged particles a good match for combination with immunotherapy? Current knowledge and perspectives. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 376:1-36. [PMID: 36997266 DOI: 10.1016/bs.ircmb.2023.01.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Charged particle radiotherapy, mainly using protons and carbon ions, provides physical characteristics allowing for a volume conformal irradiation and a reduction of the integral dose to normal tissue. Carbon ion therapy additionally features an increased biological effectiveness resulting in peculiar molecular effects. Immunotherapy, mostly performed with immune checkpoint inhibitors, is nowadays considered a pillar in cancer therapy. Based on the advantageous features of charged particle radiotherapy, we review pre-clinical evidence revealing a strong potential of its combination with immunotherapy. We argue that the combination therapy deserves further investigation with the aim of translation in clinics, where a few studies have been set up already.
Collapse
Affiliation(s)
- A Helm
- Biophysics Department, GSI, Darmstadt, Germany
| | - C Totis
- Biophysics Department, GSI, Darmstadt, Germany
| | - M Durante
- Biophysics Department, GSI, Darmstadt, Germany.
| | - C Fournier
- Biophysics Department, GSI, Darmstadt, Germany
| |
Collapse
|
12
|
Lohberger B, Barna S, Glänzer D, Eck N, Kerschbaum-Gruber S, Stasny K, Leithner A, Georg D. Cellular and Molecular Biological Alterations after Photon, Proton, and Carbon Ions Irradiation in Human Chondrosarcoma Cells Linked with High-Quality Physics Data. Int J Mol Sci 2022; 23:11464. [PMID: 36232764 PMCID: PMC9569755 DOI: 10.3390/ijms231911464] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/08/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Chondrosarcomas are particularly difficult to treat due to their resistance to chemotherapy and radiotherapy. However, particle therapy can enhance local control and patient survival rates. To improve our understanding of the basic cellular radiation response, as a function of dose and linear energy transfer (LET), we developed a novel water phantom-based setup for cell culture experiments and characterized it dosimetrically. In a direct comparison, human chondrosarcoma cell lines were analyzed with regard to their viability, cell proliferation, cell cycle, and DNA repair behavior after irradiation with X-ray, proton, and carbon ions. Our results clearly showed that cell viability and proliferation were inhibited according to the increasing ionization density, i.e., LET, of the irradiation modes. Furthermore, a prominent G2/M arrest was shown. Gene expression profiling proved the upregulation of the senescence genes CDKN1A (p21), CDKN2A (p16NK4a), BMI1, and FOXO4 after particle irradiation. Both proton or C-ion irradiation caused a positive regulation of the repair genes ATM, NBN, ATXR, and XPC, and a highly significant increase in XRCC1/2/3, ERCC1, XPC, and PCNA expression, with C-ions appearing to activate DNA repair mechanisms more effectively. The link between the physical data and the cellular responses is an important contribution to the improvement of the treatment system.
Collapse
Affiliation(s)
- Birgit Lohberger
- Department of Orthopaedics and Trauma, Medical University of Graz, 8036 Graz, Austria
| | - Sandra Barna
- Department of Radiation Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Dietmar Glänzer
- Department of Orthopaedics and Trauma, Medical University of Graz, 8036 Graz, Austria
| | - Nicole Eck
- Department of Orthopaedics and Trauma, Medical University of Graz, 8036 Graz, Austria
| | | | | | - Andreas Leithner
- Department of Orthopaedics and Trauma, Medical University of Graz, 8036 Graz, Austria
| | - Dietmar Georg
- Department of Radiation Oncology, Medical University of Vienna, 1090 Vienna, Austria
- MedAustron Ion Therapy Center, 2700 Wiener Neustadt, Austria
| |
Collapse
|
13
|
Li Y, Li X, Yang J, Wang S, Tang M, Xia J, Gao Y. Flourish of Proton and Carbon Ion Radiotherapy in China. Front Oncol 2022; 12:819905. [PMID: 35237518 PMCID: PMC8882681 DOI: 10.3389/fonc.2022.819905] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
Proton and heavy ion therapy offer superior relative biological effectiveness (RBE) in the treatment of deep-seated tumors compared with conventional photon radiotherapy due to its Bragg-peak feature of energy deposition in organs. Many proton and carbon ion therapy centers are active all over the world. At present, five particle radiotherapy institutes have been built and are receiving patient in China, mainly including Wanjie Proton Therapy Center (WPTC), Shanghai Proton Heavy Ion Center (SPHIC), Heavy Ion Cancer Treatment Center (HIMM), Chang Gung Memorial Hospital (CGMH), and Ruijin Hospital affiliated with Jiao Tong University. Many cancer patients have benefited from ion therapy, showing unique advantages over surgery and chemotherapy. By the end of 2020, nearly 8,000 patients had been treated with proton, carbon ion or carbon ion combined with proton therapy. So far, there is no systemic review for proton and carbon ion therapy facility and clinical outcome in China. We reviewed the development of proton and heavy ion therapy, as well as providing the representative clinical data and future directions for particle therapy in China. It has important guiding significance for the design and construction of new particle therapy center and patients’ choice of treatment equipment.
Collapse
Affiliation(s)
- Yue Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- *Correspondence: Yue Li,
| | - Xiaoman Li
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jiancheng Yang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Sicheng Wang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Meitang Tang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Jiawen Xia
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- Huizhou Research Center of Ion Science, Chinese Academy of Sciences, Huizhou, China
| | - Yunzhe Gao
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
- School of Nuclear Science and Technology, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
14
|
Askar MA, Thabet NM, El-Sayyad GS, El-Batal AI, Abd Elkodous M, El Shawi OE, Helal H, Abdel-Rafei MK. Dual Hyaluronic Acid and Folic Acid Targeting pH-Sensitive Multifunctional 2DG@DCA@MgO-Nano-Core-Shell-Radiosensitizer for Breast Cancer Therapy. Cancers (Basel) 2021; 13:cancers13215571. [PMID: 34771733 PMCID: PMC8583154 DOI: 10.3390/cancers13215571] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 01/16/2023] Open
Abstract
Simple Summary In this study, we have developed CD44 and folate receptor-targeting multi-functional dual drug-loaded nanoparticles. This comprises hyaluronic acid (HA) and folic acid (FA) conjugated to 2-deoxy glucose (2DG) and a shell linked to a dichloroacetate (DCA) and magnesium oxide (MgO) core (2DG@DCA@MgO; DDM) to enhance the localized chemo-radiotherapy for effective breast cancer (BC) treatment. The physicochemical properties of nanoparticles including stability, selectivity, responsive release to pH, cellular uptake, and anticancer efficacy were comprehensively examined. Mechanistically, we identified multiple component signal pathways as important regulators of BC metabolism and mediators for the inhibitory effects exerted by DDM. Nanoparticles exhibited sustained DDM release properties in bio-relevant media, which was responsive to acidic pH providing edibility to the control of drug release from nanoparticles. DDM-loaded and HA–FA-functionalized nanoparticles exhibited increased selectivity and uptake by BC cells. Cell-based assays indicated that the functionalized DDM significantly suppressed cancer cell growth and boosted radiotherapy (RT) efficacy via inducing cell cycle arrest, enhancing apoptosis, and modulating glycolytic and OXPHOS pathways. Accordingly, the inhibition of glycolysis/OXPHOS by DDM and RT treatment may result in cancer metabolic reprogramming via a novel PI3K/AKT/mTOR/P53NF-κB/VEGF pathway in BC cells. Therefore, the dual targeting of glycolysis/OXPHOS pathways is suggested as a promising antitumor strategy. Abstract Globally, breast cancer (BC) poses a serious public health risk. The disease exhibits a complex heterogeneous etiology and is associated with a glycolytic and oxidative phosphorylation (OXPHOS) metabolic reprogramming phenotype, which fuels proliferation and progression. Due to the late manifestation of symptoms, rigorous treatment regimens are required following diagnosis. Existing treatments are limited by a lack of specificity, systemic toxicity, temporary remission, and radio-resistance in BC. In this study, we have developed CD44 and folate receptor-targeting multi-functional dual drug-loaded nanoparticles. This composed of hyaluronic acid (HA) and folic acid (FA) conjugated to a 2-deoxy glucose (2DG) shell linked to a layer of dichloroacetate (DCA) and a magnesium oxide (MgO) core (2DG@DCA@MgO; DDM) to enhance the localized chemo-radiotherapy for effective BC treatment. The physicochemical properties of nanoparticles including stability, selectivity, responsive release to pH, cellular uptake, and anticancer efficacy were thoroughly examined. Mechanistically, we identified multiple component signaling pathways as important regulators of BC metabolism and mediators for the inhibitory effects elicited by DDM. Nanoparticles exhibited sustained DDM release properties in a bio-relevant media, which was responsive to the acidic pH enabling eligibility to the control of drug release from nanoparticles. DDM-loaded and HA–FA-functionalized nanoparticles exhibited increased selectivity and uptake by BC cells. Cell-based assays revealed that the functionalized DDM significantly suppressed cancer cell growth and improved radiotherapy (RT) through inducing cell cycle arrest, enhancing apoptosis, and modulating glycolytic and OXPHOS pathways. By highlighting DDM mechanisms as an antitumor and radio-sensitizing reagent, our data suggest that glycolytic and OXPHOS pathway modulation occurs via the PI3K/AKT/mTOR/NF-κB/VEGFlow and P53high signaling pathway. In conclusion, the multi-functionalized DDM opposed tumor-associated metabolic reprogramming via multiple signaling pathways in BC cells as a promising targeted metabolic approach.
Collapse
Affiliation(s)
- Mostafa A. Askar
- Radiation Biology Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo 11787, Egypt; (N.M.T.); (M.K.A.-R.)
- Correspondence: (M.A.A.); (G.S.E.-S.); Tel.: +20-010-1704-8253 (M.A.A.)
| | - Noura M. Thabet
- Radiation Biology Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo 11787, Egypt; (N.M.T.); (M.K.A.-R.)
| | - Gharieb S. El-Sayyad
- Drug Microbiology Laboratory, Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo 11787, Egypt;
- Correspondence: (M.A.A.); (G.S.E.-S.); Tel.: +20-010-1704-8253 (M.A.A.)
| | - Ahmed I. El-Batal
- Drug Microbiology Laboratory, Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo 11787, Egypt;
| | - Mohamed Abd Elkodous
- Department of Electrical and Electronic Information Engineering, Toyohashi University of Technology, Toyohashi 441-8580, Japan;
| | - Omama E. El Shawi
- Health and Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo 11787, Egypt;
| | - Hamed Helal
- Zoology Department, Faculty of Science, Al-Azhar University, Cairo 11651, Egypt;
| | - Mohamed K. Abdel-Rafei
- Radiation Biology Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo 11787, Egypt; (N.M.T.); (M.K.A.-R.)
| |
Collapse
|
15
|
Averbeck D, Rodriguez-Lafrasse C. Role of Mitochondria in Radiation Responses: Epigenetic, Metabolic, and Signaling Impacts. Int J Mol Sci 2021; 22:ijms222011047. [PMID: 34681703 PMCID: PMC8541263 DOI: 10.3390/ijms222011047] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/24/2021] [Accepted: 10/08/2021] [Indexed: 12/15/2022] Open
Abstract
Until recently, radiation effects have been considered to be mainly due to nuclear DNA damage and their management by repair mechanisms. However, molecular biology studies reveal that the outcomes of exposures to ionizing radiation (IR) highly depend on activation and regulation through other molecular components of organelles that determine cell survival and proliferation capacities. As typical epigenetic-regulated organelles and central power stations of cells, mitochondria play an important pivotal role in those responses. They direct cellular metabolism, energy supply and homeostasis as well as radiation-induced signaling, cell death, and immunological responses. This review is focused on how energy, dose and quality of IR affect mitochondria-dependent epigenetic and functional control at the cellular and tissue level. Low-dose radiation effects on mitochondria appear to be associated with epigenetic and non-targeted effects involved in genomic instability and adaptive responses, whereas high-dose radiation effects (>1 Gy) concern therapeutic effects of radiation and long-term outcomes involving mitochondria-mediated innate and adaptive immune responses. Both effects depend on radiation quality. For example, the increased efficacy of high linear energy transfer particle radiotherapy, e.g., C-ion radiotherapy, relies on the reduction of anastasis, enhanced mitochondria-mediated apoptosis and immunogenic (antitumor) responses.
Collapse
Affiliation(s)
- Dietrich Averbeck
- Laboratory of Cellular and Molecular Radiobiology, PRISME, UMR CNRS 5822/IN2P3, IP2I, Lyon-Sud Medical School, University Lyon 1, 69921 Oullins, France;
- Correspondence:
| | - Claire Rodriguez-Lafrasse
- Laboratory of Cellular and Molecular Radiobiology, PRISME, UMR CNRS 5822/IN2P3, IP2I, Lyon-Sud Medical School, University Lyon 1, 69921 Oullins, France;
- Department of Biochemistry and Molecular Biology, Lyon-Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France
| |
Collapse
|
16
|
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
|