1
|
Yang T, Gao R, Gao Y, Huang M, Cui J, Lin L, Cheng H, Dang W, Gao Y, Ma Z. The Changes of Lymphocytes and Immune Molecules in Irradiated Mice by Different Doses of Radiation. HEALTH PHYSICS 2025:00004032-990000000-00227. [PMID: 39888326 DOI: 10.1097/hp.0000000000001957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
ABSTRACT The effects of different radiation doses on T and B lymphocyte functional subsets and the changes of immune cells and immune molecules were observed in mice at different times post-irradiation to provide a theoretical basis for the changes of immune cells affected by radiation. In this study, the changes of T and B immune cells and immune-related molecules were observed at 1, 3, 7, 14, and 21 d after single irradiation of 2 Gy, 4 Gy, and 6 Gy. The results showed that white blood cells (WBC), lymphocytes (LYMPH), and lymphocyte percentage (LYMPH%) in peripheral blood of mice were significantly reduced and reached the lowest point 3 d after irradiation. Flow cytometry results showed that the percentages of CD3+T and CD8+/CD3+T lymphocytes in spleen and thymus were significantly decreased, and the percentages of CD19+B lymphocytes in spleen and CD4+/CD3+T lymphocytes in thymus were also decreased. However, the percentages of splenic NK cells, CD4+/CD3+T cells, and CD4+/CD8+ ratios in spleen and thymus were increased. Most of the indicators fell to the lowest or highest point 3 d after irradiation, indicating that immune function was suppressed at this time. From 7 to 21 d after irradiation, most immune cells gradually recovered. Single irradiation of 2 Gy, 4 Gy, and 6 Gy increased the contents of IL-1β, IL-2, IL-6, IL-17, TNF-α, TGF-β, and IFN-γ in serum of mice and decreased the contents of anti-inflammatory factors IL-4 and IL-10. The serum levels of immunoglobulin IgA, IgG, IgM and complement C3, C4 were significantly increased after irradiation. Our study showed that a single dose of 2 Gy, 4 Gy, and 6 Gy induced immunosuppression in mice, and maximum immunosuppression was achieved 3 d after irradiation. At this time, CD19+B lymphocytes were the most sensitive, followed by CD3+T lymphocytes, and NK cells were the most resistant. The radiosensitivity of CD8+/CD3+T lymphocytes was slightly higher than that of CD4+/CD3+T lymphocytes.
Collapse
Affiliation(s)
| | | | | | - Mingyue Huang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | | | | | | | | | - Yue Gao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | | |
Collapse
|
2
|
Georgescu A, Oved JH, Galarraga JH, Cantrell T, Mehta S, Dulmovits BM, Olson TS, Fattahi P, Wang A, Candarlioglu PL, Muvaffak A, Kim MM, Aydin SA, Seo J, Diffenderfer ES, Lynch A, Worthen GS, Huh DD. Self-organization of the hematopoietic vascular niche and emergent innate immunity on a chip. Cell Stem Cell 2024; 31:1847-1864.e6. [PMID: 39642865 DOI: 10.1016/j.stem.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 07/04/2024] [Accepted: 11/05/2024] [Indexed: 12/09/2024]
Abstract
Here, we present a bioengineering approach to emulate the human bone marrow in vitro. Our developmentally inspired method uses self-organization of human hematopoietic stem and progenitor cells and vascular endothelial cells cultured in a three-dimensional microphysiological system to create vascularized, perfusable tissue constructs that resemble the hematopoietic vascular niche of the human marrow. The microengineered niche is capable of multilineage hematopoiesis and can generate functionally mature human myeloid cells that can intravasate into perfused blood vessels, providing a means to model the mobilization of innate immune cells from the marrow. We demonstrate the application of this system by presenting a specialized model of ionizing radiation-induced bone marrow injury and a multiorgan model of acute innate immune responses to bacterial lung infection. Furthermore, we introduce an advanced platform that enables large-scale integration and automated experimentation of the engineered hematopoietic tissues for preclinical screening of myelotoxicity due to anti-cancer drugs.
Collapse
Affiliation(s)
- Andrei Georgescu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Vivodyne Inc., Philadelphia, PA 19104, USA
| | - Joseph Hai Oved
- Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | - Samira Mehta
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brian M Dulmovits
- Blood and Marrow Transplant Program, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Timothy S Olson
- Blood and Marrow Transplant Program, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Pouria Fattahi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anni Wang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Asli Muvaffak
- Complex In Vitro Models, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Michele M Kim
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sezin Aday Aydin
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jeongyun Seo
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eric S Diffenderfer
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anthony Lynch
- Complex In Vitro Models, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - G Scott Worthen
- Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, PA 19104, USA.
| | - Dan Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Vivodyne Inc., Philadelphia, PA 19104, USA; Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
3
|
Zheng C, Yu L, Jiang Y. Radiation-induced rhinosinusitis: Mechanism research and clinical progress review. World J Otorhinolaryngol Head Neck Surg 2024; 10:324-332. [PMID: 39677057 PMCID: PMC11634722 DOI: 10.1002/wjo2.134] [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: 05/24/2023] [Revised: 07/08/2023] [Accepted: 07/20/2023] [Indexed: 12/17/2024] Open
Abstract
Objectives Radiation-induced rhinosinusitis is a vital dose-limiting reaction in patients with head and neck malignancy. Unlike oral mucositis during or after radiotherapy, radiation-induced sinusitis is easily overlooked in clinical practice and rarely included in experimental studies. Herein, we review the literature to date on radiation-induced rhinosinusitis. Methods Relevant studies published between 1995 and 2022 were determined through a detailed search using open keywords from PubMed, with manual search of the reference list of the identified articles. Keywords searched were "ionizing radiation," "radiotherapy," "intensity-modulated radiotherapy," "head and neck tumor," "nasopharyngeal carcinoma," "nasal epithelium," "radiation damage," and "radiation-induced rhinosinusitis." Full-text articles that clearly stated the pathogenesis, clinical manifestation, predictors, treatment, and prognosis of radiation-induced rhinosinusitis were included. Results Radiation-induced rhinosinusitis occurs during radiotherapy and can last for months or even years after radiotherapy. A mixture of cellular outcomes caused by ionizing radiation and persistent damage of the epithelial and submucosal tissues after the treatment result from the radiotherapy itself. Endoscopic sinus surgery improves symptoms but can be accompanied by intraoperative and postoperative complications. Nasal irrigation, steroids, and antibiotics appear to reduce inflammation and relieve symptoms to a certain extent. Studies on other potentially useful drugs are underway and in the exploration stage, without clinical application. Conclusions Despite its high incidence, radiation-induced rhinosinusitis is a type of dose-limiting toxicity that theoretically does not produce fatal effects at controlled doses and with adequate follow-up care. In moderate-to-severe cases, toxicity may be present. Currently, radiation-induced rhinosinusitis has potential prevention and treatment strategies. However, no unified management protocol has shown significant improvement in radiation-induced rhinosinusitis. Further research is necessary.
Collapse
Affiliation(s)
- Chunge Zheng
- Department of Otolaryngology, Head and Neck SurgeryThe Affiliated Hospital of Qingdao UniversityQingdaoShandong ProvinceChina
| | - Longgang Yu
- Otorhinolaryngology Head and Neck Surgery Key Laboratory of Shandong ProvinceQingdaoShandong ProvinceChina
| | - Yan Jiang
- Department of Otolaryngology, Head and Neck SurgeryThe Affiliated Hospital of Qingdao UniversityQingdaoShandong ProvinceChina
| |
Collapse
|
4
|
Nikiforov VS, Kotikova AI, Blinova EA, Akleyev AV. Transcriptional Activity of Genes Regulating T-Helper Differentiation in the Accidentally Exposed Population of the Southern Urals. DOKL BIOCHEM BIOPHYS 2024; 519:499-505. [PMID: 39283554 DOI: 10.1134/s1607672924701114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 01/19/2025]
Abstract
The objective of this work was to study the expression of the TBX21, RORC, GATA3, NFKB1, MAPK8, and STAT3 genes responsible for the regulation of the differentiation of various T-helper subpopulations in individuals chronically exposed to radiation. The object of the study was peripheral blood cells obtained from 120 persons chronically exposed to radiation in a wide range of doses on the Techa River. The mean cumulative absorbed dose to red bone marrow in the examined exposed individuals was 742.7 ± 78.6 mGy (dose range, 73.5-3516.1 mGy); in the comparison group, 17.4 ± 2.2 mGy (dose range, 0.0-55.5 mGy). The subpopulation composition of T-helpers (Th1, Th2, and Th17) was analyzed by flow cytofluorometry. The relative mRNA content of the TBX21, RORC, GATA3, NFKB1, MAPK8, and STAT3 genes was estimated by real-time PCR. The study made it possible to note a decrease in the relative number of T-helpers 2 in the populations of T-helpers of the central memory in the group of chronically exposed persons compared to the comparison group. In the population of T-helpers of the central memory, a statistically significant increase in the relative number of T-helpers 1 was shown, depending on the accumulated absorbed dose to red bone marrow. No changes in mRNA expression of the studied genes were observed. The analysis of the correlation between the expression of GATA3, MAPK8, STAT3, RORC, and TBX21 mRNA and the relative number of cells in subpopulations of T-helper types 1, 2, and 17 in the examined people did not reveal statistically significant patterns.
Collapse
Affiliation(s)
- V S Nikiforov
- Urals Research Center for Radiation Medicine of the Federal Medical and Biological Agency of Russia, Chelyabinsk, Russia.
- Chelyabinsk State University, Chelyabinsk, Russia.
| | - A I Kotikova
- Urals Research Center for Radiation Medicine of the Federal Medical and Biological Agency of Russia, Chelyabinsk, Russia
- Chelyabinsk State University, Chelyabinsk, Russia
| | - E A Blinova
- Urals Research Center for Radiation Medicine of the Federal Medical and Biological Agency of Russia, Chelyabinsk, Russia
- Chelyabinsk State University, Chelyabinsk, Russia
| | - A V Akleyev
- Urals Research Center for Radiation Medicine of the Federal Medical and Biological Agency of Russia, Chelyabinsk, Russia
- Chelyabinsk State University, Chelyabinsk, Russia
| |
Collapse
|
5
|
Miranda S, Vermeesen R, Janssen A, Rehnberg E, Etlioglu E, Baatout S, Tabury K, Baselet B. Effects of simulated space conditions on CD4+ T cells: a multi modal analysis. Front Immunol 2024; 15:1443936. [PMID: 39286254 PMCID: PMC11402665 DOI: 10.3389/fimmu.2024.1443936] [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: 06/04/2024] [Accepted: 08/08/2024] [Indexed: 09/19/2024] Open
Abstract
Introduction The immune system is an intricate network of cellular components that safeguards against pathogens and aberrant cells, with CD4+ T cells playing a central role in this process. Human space travel presents unique health challenges, such as heavy ion ionizing radiation, microgravity, and psychological stress, which can collectively impede immune function. The aim of this research was to examine the consequences of simulated space stressors on CD4+ T cell activation, cytokine production, and gene expression. Methods CD4+ T cells were obtained from healthy individuals and subjected to Fe ion particle radiation, Photon irradiation, simulated microgravity, and hydrocortisone, either individually or in different combinations. Cytokine levels for Th1 and Th2 cells were determined using multiplex Luminex assays, and RNA sequencing was used to investigate gene expression patterns and identify essential genes and pathways impacted by these stressors. Results Simulated microgravity exposure resulted in an apparent Th1 to Th2 shift, evidenced on the level of cytokine secretion as well as altered gene expression. RNA sequencing analysis showed that several gene pathways were altered, particularly in response to Fe ions irradiation and simulated microgravity exposures. Individually, each space stressor caused differential gene expression, while the combination of stressors revealed complex interactions. Discussion The research findings underscore the substantial influence of the space exposome on immune function, particularly in the regulation of T cell responses. Future work should focus expanding the limited knowledge in this field. Comprehending these modifications will be essential for devising effective strategies to safeguard the health of astronauts during extended space missions. Conclusion The effects of simulated space stressors on CD4+ T cell function are substantial, implying that space travel poses a potential threat to immune health. Additional research is necessary to investigate the intricate relationship between space stressors and to develop effective countermeasures to mitigate these consequences.
Collapse
Affiliation(s)
- Silvana Miranda
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Randy Vermeesen
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
| | - Ann Janssen
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
| | - Emil Rehnberg
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Emre Etlioglu
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Kevin Tabury
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
- Department of Biomedical Engineering, College of Engineering and Computing, University of South Carolina, Columbia, SC, United States
| | - Bjorn Baselet
- Radiobiology Unit, Institute for Nuclear Medical Applications, Belgian Nuclear Research Centre SCK CEN, Mol, Belgium
| |
Collapse
|
6
|
McDowell JA, Kosmacek EA, Baine MJ, Adebisi O, Zheng C, Bierman MM, Myers MS, Chatterjee A, Liermann-Wooldrik KT, Lim A, Dickinson KA, Oberley-Deegan RE. Exogenous APN protects normal tissues from radiation-induced oxidative damage and fibrosis in mice and prostate cancer patients with higher levels of APN have less radiation-induced toxicities. Redox Biol 2024; 73:103219. [PMID: 38851001 PMCID: PMC11201354 DOI: 10.1016/j.redox.2024.103219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/26/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024] Open
Abstract
Radiation causes damage to normal tissues that leads to increased oxidative stress, inflammation, and fibrosis, highlighting the need for the selective radioprotection of healthy tissues without hindering radiotherapy effectiveness in cancer. This study shows that adiponectin, an adipokine secreted by adipocytes, protects normal tissues from radiation damage invitro and invivo. Specifically, adiponectin (APN) reduces chronic oxidative stress and fibrosis in irradiated mice. Importantly, APN also conferred no protection from radiation to prostate cancer cells. Adipose tissue is the primary source of circulating endogenous adiponectin. However, this study shows that adipose tissue is sensitive to radiation exposure exhibiting morphological changes and persistent oxidative damage. In addition, radiation results in a significant and chronic reduction in blood APN levels from adipose tissue in mice and human prostate cancer patients exposed to pelvic irradiation. APN levels negatively correlated with bowel toxicity and overall toxicities associated with radiotherapy in prostate cancer patients. Thus, protecting, or modulating APN signaling may improve outcomes for prostate cancer patients undergoing radiotherapy.
Collapse
Affiliation(s)
- Joshua A McDowell
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Elizabeth A Kosmacek
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Michael J Baine
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Oluwaseun Adebisi
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Cheng Zheng
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Madison M Bierman
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Molly S Myers
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Arpita Chatterjee
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kia T Liermann-Wooldrik
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Andrew Lim
- College of Nursing, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kristin A Dickinson
- College of Nursing, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Rebecca E Oberley-Deegan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
7
|
Masoudi S, Kalani M, Alavianmehr A, Mosleh-Shirazi MA, Mortazavi SMJ, Farjadian S. Sequential radiation exposure: uncovering the potential of low dose ionizing radiation in mitigating high dose effects on immune cells. Int J Radiat Biol 2024; 100:1009-1018. [PMID: 38776451 DOI: 10.1080/09553002.2024.2345107] [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: 09/18/2023] [Accepted: 04/09/2024] [Indexed: 05/25/2024]
Abstract
PURPOSE The radioadaptive response refers to a phenomenon wherein exposure to a low dose of ionizing radiation (LDIR) can induce a protective response in cells or organisms, reducing the adverse effects of a subsequent higher dose of ionizing radiation (HDIR). However, it is possible to administer the low dose after the challenge dose. This study was conducted to determine the potential mitigating effect of LDIR administered after HDIR on mice immune cells. MATERIALS AND METHODS Alongside the conventional adaptive response setting, one group of mice was initially exposed to HDIR and subsequently treated with LDIR. Neutrophil activation was done using DHR-reducing assay and cell proliferation was evaluated through CFSE-dilution assay in helper (CD4+) and cytotoxic (CD8+) T cells. Cytokine production by these T cell subsets was also assessed by intracellular staining using flow cytometry. RESULTS The results of this study revealed no change in neutrophil function between any of the mice groups compared to the untreated control group. Although significant changes were not detected in the proliferation of CD4+ T cells, decreased proliferation was observed in stimulated CD8+ T cells in the HDIR group. In contrast to IFN-ɣ, which showed no evident change in either of the T cell subsets after stimulation, IL-4 was rigorously dropped in stimulated CD4+ T cells in the HDIR group. CONCLUSIONS In summary, the results of this study indicated that the administration of LDIR to mice before HDIR was not able to reduce the detrimental effects of HDIR in our experimental setting. Instead, we observed a mitigating effect of LDIR when administered after the challenge dose. This suggests that not only the dose and duration but also the order of LDIR relative to HDIR affects its efficacy.
Collapse
Affiliation(s)
- Sadegh Masoudi
- The Ionizing and Non-Ionizing Radiation Protection Research Center (INIRPRC), School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Kalani
- Professor Alborzi Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Alavianmehr
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Amin Mosleh-Shirazi
- The Ionizing and Non-Ionizing Radiation Protection Research Center (INIRPRC), School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- Radiotherapy Department and Center for Research in Medical Physics and Biomedical Engineering, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Javad Mortazavi
- The Ionizing and Non-Ionizing Radiation Protection Research Center (INIRPRC), School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medical Physics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shirin Farjadian
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
8
|
Chen YY, Wang M, Zuo CY, Mao MX, Peng XC, Cai J. Nrf-2 as a novel target in radiation induced lung injury. Heliyon 2024; 10:e29492. [PMID: 38665580 PMCID: PMC11043957 DOI: 10.1016/j.heliyon.2024.e29492] [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: 10/16/2023] [Revised: 03/09/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Radiation-induced lung injury (RILI) is a common and fatal complication of chest radiotherapy. The underlying mechanisms include radiation-induced oxidative stress caused by damage to the deoxyribonucleic acid (DNA) and production of reactive oxygen species (ROS), resulting in apoptosis of lung and endothelial cells and recruitment of inflammatory cells and myofibroblasts expressing NADPH oxidase to the site of injury, which in turn contribute to oxidative stress and cytokine production. Nuclear factor erythroid 2-related factor 2 (Nrf-2) is a vital transcription factor that regulates oxidative stress and inhibits inflammation. Studies have shown that Nrf-2 protects against radiation-induced lung inflammation and fibrosis. This review discusses the protective role of Nrf-2 in RILI and its possible mechanisms.
Collapse
Affiliation(s)
- Yuan-Yuan Chen
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, 434023, PR China
| | - Meng Wang
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, 434023, PR China
| | - Chen-Yang Zuo
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, 434023, PR China
| | - Meng-Xia Mao
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, 434023, PR China
| | - Xiao-Chun Peng
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, 434023, PR China
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, 434023, PR China
| | - Jun Cai
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, 434023, PR China
| |
Collapse
|
9
|
Liu S, Wang W, Hu S, Jia B, Tuo B, Sun H, Wang Q, Liu Y, Sun Z. Radiotherapy remodels the tumor microenvironment for enhancing immunotherapeutic sensitivity. Cell Death Dis 2023; 14:679. [PMID: 37833255 PMCID: PMC10575861 DOI: 10.1038/s41419-023-06211-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/22/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023]
Abstract
Cancer immunotherapy has transformed traditional treatments, with immune checkpoint blockade being particularly prominent. However, immunotherapy has minimal benefit for patients in most types of cancer and is largely ineffective in some cancers (such as pancreatic cancer and glioma). A synergistic anti-tumor response may be produced through the combined application with traditional tumor treatment methods. Radiotherapy (RT) not only kills tumor cells but also triggers the pro-inflammatory molecules' release and immune cell infiltration, which remodel the tumor microenvironment (TME). Therefore, the combination of RT and immunotherapy is expected to achieve improved efficacy. In this review, we summarize the effects of RT on cellular components of the TME, including T cell receptor repertoires, different T cell subsets, metabolism, tumor-associated macrophages and other myeloid cells (dendritic cells, myeloid-derived suppressor cells, neutrophils and eosinophils). Meanwhile, non-cellular components such as lactate and extracellular vesicles are also elaborated. In addition, we discuss the impact of different RT modalities on tumor immunity and issues related to the clinical practice of combination therapy.
Collapse
Affiliation(s)
- Senbo Liu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Wenkang Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Shengyun Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Bin Jia
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Baojing Tuo
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Haifeng Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Qiming Wang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 450001, Zhengzhou, China.
| | - Yang Liu
- Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 450001, Zhengzhou, China.
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
| |
Collapse
|
10
|
Ebrahimi HA, Larizadeh MH, Saba M, Jafarzadeh A. Radiotherapy Improves the Disability in Patients with Secondary Progressive Multiple Sclerosis. J Biomed Phys Eng 2023; 13:317-322. [PMID: 37609511 PMCID: PMC10440411 DOI: 10.31661/jbpe.v0i0.2012-1238] [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: 12/03/2020] [Accepted: 04/28/2021] [Indexed: 08/24/2023]
Abstract
Background Multiple sclerosis (MS) as a complex neurological abnormality is marked with loss of myelin and axons due to chronic inflammatory and autoimmune responses. The modulatory properties of the low dose radiation (LDR) on inflammatory and immune responses have well known. Objective The current research aimed to assess the impacts of LDR on the disability in patients suffering from MS. Material and Methods This experimental pilot study was done on 10 patients with secondary progressive multiple sclerosis (SPMS). After magnetic resonance imaging, the SPMS patients were treated by LDR at a daily dose of 2 Gray for 5 consecutive days (totally 10 Gray dose) using a linear accelerator. The extent of the disability was evaluated one week after the completion of radiotherapy using expanded disability status scale (EDSS). Results After receiving radiotherapy, the patients had a feeling of wellbeing of some sort. The mean of EDSS was significantly reduced after radiotherapy compared with before irradiation (7.4±0.45 vs 6.35±1.18; P<0.017). EDSS more decreased in younger SPMS patients (P=0.0001), and in the women after LDR (P=0.027). Conclusion Radiotherapy can reduce fatigue and EDSS in patients with SPMS. The age and gender of patients may influence the LDR efficacy.
Collapse
Affiliation(s)
- Hossein-Ali Ebrahimi
- Neurology Research Center, Department of Neurology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad-Hasan Larizadeh
- Neurology Research Center, Department of Neurology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Saba
- Department of Radiology, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Abdollah Jafarzadeh
- Department of Immunology, Medical School, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Immunology, Medical School, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
11
|
Hughes R, Snook AE, Mueller AC. The poorly immunogenic tumor microenvironment of pancreatic cancer: the impact of radiation therapy, and strategies targeting resistance. Immunotherapy 2022; 14:1393-1405. [PMID: 36468417 DOI: 10.2217/imt-2022-0046] [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: 12/12/2022] Open
Abstract
Pancreatic cancer is one of the most lethal cancers, due to its uniquely aggressive behavior and resistance to therapy. The tumor microenvironment of pancreatic cancer is immunosuppressive, and attempts at utilizing immunotherapies have been unsuccessful. Radiation therapy (RT) results in immune activation and antigen presentation in other cancers, but in pancreatic cancer has had limited success in stimulating immune responses. RT activates common pathways of fibrosis and chronic inflammation seen in pancreatic cancer, resulting in immune suppression. Here we describe the pancreatic tumor microenvironment with regard to fibrosis, myeloid and lymphoid cells, and the impact of RT. We also describe strategies of targeting these pathways that have promise to improve outcomes by harnessing the cytotoxic and immune-activating aspects of RT.
Collapse
Affiliation(s)
- Robert Hughes
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Adam E Snook
- Department of Pharmacology & Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA.,Department of Microbiology & Immunology, Thomas Jefferson University, Philadelphia, PA, USA; Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Adam C Mueller
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| |
Collapse
|
12
|
Askar MA, Guida MS, AbuNour SM, Ragab EA, Ali EN, Abdel-Magied N, Mansour NA, Elmasry SA. Nanoparticles for active combination radio mitigating agents of zinc coumarate and zinc caffeinate in a rat model. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:30233-30248. [PMID: 35000180 DOI: 10.1007/s11356-021-18411-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 12/26/2021] [Indexed: 06/14/2023]
Abstract
Zinc coumarate and zinc caffeinate nanoparticles (ZnCoNPs, ZnCaNPs) affect different biological processes. This study aimed to evaluate the mitigating action of ZnCoNPs in combination with ZnCaNPs against liver damage induced by gamma rays (γ-rays). Rats were exposed to 7 Gy of γ-rays and then injected intraperitoneally (i.p) with ZnCoNPs [2U/rat/day (5 mg/kg)] and ZnCaNPs [2U/rat/day (15 mg/kg)] for 7 consecutive days. The results showed that irradiated rats treated with ZnCoNPs (5 mg/kg/body weight) in combination with ZnCaNPs (15 mg/kg/body weight) for 7 days had a significant increases in body weight, antioxidant levels, T helper cell 4 (cluster of differentiation 4 (CD4)), and T cytotoxic cell 8 (cluster of differentiation 8 (CD8)), associated with a marked decrease in lipid peroxidation (LP), nitric oxide(NOx), total free radicals concentrate (TFRC), and DNA fragmentation. There were positive alterations in the morphological state, hematological parameters and the cell cycle phases. Additionally, the histopathological study demonstrated an improvement in the liver tissue of irradiated rats after treatment. Thus, ZnCoNPs and ZnCaNPs could be used as natural mitigating agents to reduce the hazards of ionizing radiation.
Collapse
Affiliation(s)
- Mostafa A Askar
- Department of Radiation Biology Research, National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Cairo, Egypt
| | - Mona S Guida
- GenticUnit, Pediatric Hospital, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Seham M AbuNour
- Department of Health and Radiation Research, National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Cairo, Egypt
| | - Essam A Ragab
- Department of Natural Product Research, National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Cairo, Egypt
| | - Eiman N Ali
- Department of Radiation Biology Research, National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Cairo, Egypt
| | - Nadia Abdel-Magied
- Department of Radiation Biology Research, National Centre for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Cairo, Egypt.
| | - Nahla A Mansour
- Petrochemical Department, Egyptian Petroleum Research Institute, Nasr City, Cairo, Egypt
| | - Samir A Elmasry
- Department of Molecular Biology, Genetic Engineering & Biotechnology Research Institute, Sadat City, Egypt
| |
Collapse
|
13
|
Prolonged Inhalation Exposure to Coal Dust on Irradiated Rats and Consequences. ScientificWorldJournal 2022; 2022:8824275. [PMID: 35153629 PMCID: PMC8828334 DOI: 10.1155/2022/8824275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 12/17/2021] [Accepted: 12/23/2021] [Indexed: 11/21/2022] Open
Abstract
The purposes of this study were to research immune system changes and liver and lung tissues in irradiated rats after prolonged exposure to coal dust. A study was carried out on 30 male Wistar rats that were divided into 3 groups: group I, intact animals; group II, exposure to coal dust and 0.2 Gy γ-irradiation; and group III, combined exposure to 6 Gy γ-irradiation and coal dust. The combination of a low and sublethal dose of γ-irradiation with coal dust leads to a significant change in immunity at the remote period. Particularly, the increase in radioactivity at the combined effect causes weakening of phagocytosis, and reduction in T lymphocytes by a factor of 2, immunoglobulin imbalance, and cytokine dysfunction develop secondary immune failure. During prolonged inhalation with coal dust of irradiated animals with the dose of 0.2 Gy, fibrosis and perivascular sclerosis of the bronchial wall of the lungs are formed, and perivascular fibrosis is formed in the liver. The increase in exposure dose up to 6 Gy in combination with coal, in the distant period, caused pulmonary hypertension amid hypertrophy of light arterial vessels and fibrous changes in arteriole, and destructive changes and collection necrosis develop in liver parenchyma. In the case of dust radiation synergy, the increase in doses leads to a significant immune deficiency, which occurs according to the “dose effect” principle; increases damage to animal tissues; and leads to liver tissue necrosis, pulmonary fibrosis, and pulmonary hypertension.
Collapse
|
14
|
Franzese O, Torino F, Giannetti E, Cioccoloni G, Aquino A, Faraoni I, Fuggetta MP, De Vecchis L, Giuliani A, Kaina B, Bonmassar E. Abscopal Effect and Drug-Induced Xenogenization: A Strategic Alliance in Cancer Treatment? Int J Mol Sci 2021; 22:ijms221910672. [PMID: 34639014 PMCID: PMC8509363 DOI: 10.3390/ijms221910672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/23/2021] [Accepted: 09/28/2021] [Indexed: 11/16/2022] Open
Abstract
The current state of cancer treatment is still far from being satisfactory considering the strong impairment of patients' quality of life and the high lethality of malignant diseases. Therefore, it is critical for innovative approaches to be tested in the near future. In view of the crucial role that is played by tumor immunity, the present review provides essential information on the immune-mediated effects potentially generated by the interplay between ionizing radiation and cytotoxic antitumor agents when interacting with target malignant cells. Therefore, the radiation-dependent abscopal effect (i.e., a biological effect of ionizing radiation that occurs outside the irradiated field), the influence of cancer chemotherapy on the antigenic pattern of target neoplastic cells, and the immunogenic cell death (ICD) caused by anticancer agents are the main topics of this presentation. It is widely accepted that tumor immunity plays a fundamental role in generating an abscopal effect and that anticancer drugs can profoundly influence not only the host immune responses, but also the immunogenic pattern of malignant cells. Remarkably, several anticancer drugs impact both the abscopal effect and ICD. In addition, certain classes of anticancer agents are able to amplify already expressed tumor-associated antigens (TAA). More importantly, other drugs, especially triazenes, induce the appearance of new tumor neoantigens (TNA), a phenomenon that we termed drug-induced xenogenization (DIX). The adoption of the abscopal effect is proposed as a potential therapeutic modality when properly applied concomitantly with drug-induced increase in tumor cell immunogenicity and ICD. Although little to no preclinical or clinical studies are presently available on this subject, we discuss this issue in terms of potential mechanisms and therapeutic benefits. Upcoming investigations are aimed at evaluating how chemical anticancer drugs, radiation, and immunotherapies are interacting and cooperate in evoking the abscopal effect, tumor xenogenization and ICD, paving the way for new and possibly successful approaches in cancer therapy.
Collapse
Affiliation(s)
- Ornella Franzese
- School of Medicine, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (O.F.); (G.C.); (A.A.); (I.F.); (L.D.V.)
| | - Francesco Torino
- Department of Systems Medicine, Medical Oncology, University of Rome Tor Vergata, 00133 Rome, Italy; (F.T.); (E.G.)
| | - Elisa Giannetti
- Department of Systems Medicine, Medical Oncology, University of Rome Tor Vergata, 00133 Rome, Italy; (F.T.); (E.G.)
| | - Giorgia Cioccoloni
- School of Medicine, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (O.F.); (G.C.); (A.A.); (I.F.); (L.D.V.)
- School of Food Science and Nutrition, University of Leeds, Leeds LS29JT, UK
| | - Angelo Aquino
- School of Medicine, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (O.F.); (G.C.); (A.A.); (I.F.); (L.D.V.)
| | - Isabella Faraoni
- School of Medicine, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (O.F.); (G.C.); (A.A.); (I.F.); (L.D.V.)
| | - Maria Pia Fuggetta
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Via Fosso del Cavaliere, 00133 Rome, Italy; (M.P.F.); (A.G.)
| | - Liana De Vecchis
- School of Medicine, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (O.F.); (G.C.); (A.A.); (I.F.); (L.D.V.)
| | - Anna Giuliani
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Via Fosso del Cavaliere, 00133 Rome, Italy; (M.P.F.); (A.G.)
| | - Bernd Kaina
- Institute of Toxicology, University Medical Center, D-55131 Mainz, Germany
- Correspondence: (B.K.); (E.B.)
| | - Enzo Bonmassar
- School of Medicine, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (O.F.); (G.C.); (A.A.); (I.F.); (L.D.V.)
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Via Fosso del Cavaliere, 00133 Rome, Italy; (M.P.F.); (A.G.)
- Correspondence: (B.K.); (E.B.)
| |
Collapse
|
15
|
Saleeva DV, Zasukhina GD. [Prospects for using low-dose radiation in the complex therapy for COVID-19]. Vopr Virusol 2021; 66:252-258. [PMID: 34545717 DOI: 10.36233/0507-4088-62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 09/17/2021] [Indexed: 11/05/2022]
Abstract
This review presents the literature data of new approaches for the treatment of COVID-19 with low doses of radiation (LDR). In addition, data on the use of LDR for the treatment of various disorders, in particular pneumonia, a number of inflammatory processes of various etiology, as well as Alzheimer's disease are discussed. The mechanisms of LDR action are briefly described, associated with the activation of the immune system and antiinflammatory response due to the effect on the processes of oxidative stress, which is reflected in an increase in the activity of cytokines (interleukin- (IL-) 6), changes in the expression of a number of genes (such as P53 and NF-κB (p65)) and long non-coding RNAs (ncRNAs) (the authors' own data are presented). Based on the analysis of the material presented, it can be assumed that further clinical trials of the effect of MDR (5-10 cGy) on patients with COVID-19, who are at different stages of the disease, will reveal the optimal conditions for the development and use of an effective treatment regimen.
Collapse
Affiliation(s)
- D V Saleeva
- FSBI «State Research Center - Burnasyan Federal Medical Biophysical Center» of Federal Medical Biological Agency of Russia
| | - G D Zasukhina
- FSBI «State Research Center - Burnasyan Federal Medical Biophysical Center» of Federal Medical Biological Agency of Russia; FSBIS Vavilov Institute of General Genetics of Russian Academy of Sciences
| |
Collapse
|
16
|
Choi YY, Kim A, Seong KM. Chronic radiation exposure aggravates atherosclerosis by stimulating neutrophil infiltration. Int J Radiat Biol 2021; 97:1270-1281. [PMID: 34032557 DOI: 10.1080/09553002.2021.1934750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/07/2021] [Accepted: 05/18/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Radiation exposure is known to increase the risk of chronic inflammatory diseases, such as atherosclerosis, by modulating inflammation. METHODS To investigate the infiltration of leukocytes in radiation-aggravated atherosclerosis, we examined low-density lipoprotein receptor-deficient (Ldlr-/-) mice and C57BL/6j mice after exposure to 0.5 or 1 Gy radiation over 16 weeks. RESULTS We found that radiation exposure induced atherosclerosis development in Ldlr-/- mice, as demonstrated by increased lipid-laden plaque size, reactive oxygen species levels, and levels of the pro-inflammatory cytokines, IL-1β and TNF-α, in the aortas and spleens. Total plasma cholesterol, triglyceride, and LDL cholesterol levels were also increased by radiation exposure, along with cardiovascular risk. We also showed dose-dependent increases in neutrophils and monocytes that coincided with a reduction in lymphocytes in the spleens of Ldlr-/- mice. The correlation between the infiltration of leukocytes and cytokine production was also confirmed in the hearts and spleens of these mice. CONCLUSIONS We concluded that chronic radiation exposure increased the production of pro-inflammatory mediators, which was associated with the migration of neutrophils and inflammatory monocytes into sites of atherosclerosis. Thus, our data suggest that the accumulation of neutrophils and inflammatory monocytes, together with the reduction of lymphocytes, contribute to aggravated atherosclerosis in Ldlr-/- mice under prolonged exposure to radiation.
Collapse
Affiliation(s)
- You Yeon Choi
- Laboratory of Biodosimetry, National Radiation Emergency Medical Center, KIRAMS, Seoul, Republic of Korea
| | - Areumnuri Kim
- Laboratory of Radiation Exposure and Therapeutics, National Radiation Emergency Medical Center, KIRAMS, Seoul, Republic of Korea
| | - Ki Moon Seong
- Laboratory of Biodosimetry, National Radiation Emergency Medical Center, KIRAMS, Seoul, Republic of Korea
| |
Collapse
|
17
|
Mechanisms of radiation-induced endothelium damage: Emerging models and technologies. Radiother Oncol 2021; 158:21-32. [PMID: 33581220 DOI: 10.1016/j.radonc.2021.02.007] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/24/2022]
Abstract
Radiation-induced endothelial/vascular injury is a major complicating factor in radiotherapy and a leading cause of morbidity and mortality in nuclear or radiological catastrophes. Exposure of tissue to ionizing radiation (IR) leads to the release of oxygen radicals and proteases that result in loss of endothelial barrier function and leukocyte dysfunction leading to tissue injury and organ damage. Microvascular endothelial cells are particularly sensitive to IR and radiation-induced alterations in endothelial cell function are thought to be a critical factor in organ damage through endothelial cell activation, enhanced leukocyte-endothelial cell interactions, increased barrier permeability and initiation of apoptotic pathways. These radiation-induced inflammatory responses are important in early and late radiation pathologies in various organs. A better understanding of mechanisms of radiation-induced endothelium dysfunction is therefore vital, as radiobiological response of endothelium is of major importance for medical management and therapeutic development for radiation injuries. In this review, we summarize the current knowledge of cellular and molecular mechanisms of radiation-induced endothelium damage and their impact on early and late radiation injury. Furthermore, we review established and emerging in vivo and in vitro models that have been developed to study the mechanisms of radiation-induced endothelium damage and to design, develop and rapidly screen therapeutics for treatment of radiation-induced vascular damage. Currently there are no specific therapeutics available to protect against radiation-induced loss of endothelial barrier function, leukocyte dysfunction and resulting organ damage. Developing therapeutics to prevent endothelium dysfunction and normal tissue damage during radiotherapy can serve as the urgently needed medical countermeasures.
Collapse
|
18
|
Donlon NE, Power R, Hayes C, Reynolds JV, Lysaght J. Radiotherapy, immunotherapy, and the tumour microenvironment: Turning an immunosuppressive milieu into a therapeutic opportunity. Cancer Lett 2021; 502:84-96. [PMID: 33450360 DOI: 10.1016/j.canlet.2020.12.045] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/07/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023]
Abstract
Immune checkpoint blockade (ICB) has revolutionised the treatment of solid tumours, yet most patients do not derive a clinical benefit. Resistance to ICB is often contingent on the tumour microenvironment (TME) and modulating aspects of this immunosuppressive milieu is a goal of combination treatment approaches. Radiation has been used for over a century in the management of cancer with more than half of all cancer patients receiving radiotherapy. Here, we outline the rationale behind combining radiotherapy with ICB, a potential synergy through mutually beneficial remodelling of the TME. We discuss the pleiotropic effects radiation has on the TME including immunogenic cell death, activation of cytosolic DNA sensors, remodelling the stroma and vasculature, and paradoxical infiltration of both anti-tumour and suppressive immune cell populations. These events depend on the radiation dose and fractionation and optimising these parameters will be key to develop safe and effective combination regimens. Finally, we highlight ongoing efforts that combine radiation, immunotherapy and inhibitors of DNA damage response, which can help achieve a favourable equilibrium between the immunogenic and tolerogenic effects of radiation on the immune microenvironment.
Collapse
Affiliation(s)
- N E Donlon
- Department of Surgery, School of Medicine, Trinity College Dublin, Dublin, Ireland; Trinity St James' Cancer Institute, St James's Hospital Dublin, Ireland
| | - R Power
- Department of Surgery, School of Medicine, Trinity College Dublin, Dublin, Ireland; Trinity St James' Cancer Institute, St James's Hospital Dublin, Ireland
| | - C Hayes
- Department of Surgery, School of Medicine, Trinity College Dublin, Dublin, Ireland; Trinity St James' Cancer Institute, St James's Hospital Dublin, Ireland
| | - J V Reynolds
- Department of Surgery, School of Medicine, Trinity College Dublin, Dublin, Ireland; Trinity St James' Cancer Institute, St James's Hospital Dublin, Ireland
| | - J Lysaght
- Department of Surgery, School of Medicine, Trinity College Dublin, Dublin, Ireland; Trinity St James' Cancer Institute, St James's Hospital Dublin, Ireland.
| |
Collapse
|
19
|
Pandey BN. Low-dose radiation therapy for coronavirus disease-2019 pneumonia: Is it time to look beyond apprehensions? Ann Thorac Med 2020; 15:199-207. [PMID: 33381234 PMCID: PMC7720738 DOI: 10.4103/atm.atm_433_20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 08/15/2020] [Indexed: 11/11/2022] Open
Abstract
Coronavirus disease-2019 (COVID-19) has become a global health crisis. Mortality associated with COVID-19 is characterized mainly by acute respiratory distress syndrome (ARDS), sepsis, pneumonia, and respiratory failure. The pathogenesis of the disease is known to be associated with pro-inflammatory processes after virus infection. Hence, various therapeutic strategies are being developed to control the inflammation and cytokine storm in COVID-19 patients. Recently, low-dose radiation therapy (LDRT) has been suggested for the treatment of pneumonia/ADRS in COVID-19 patients through irradiation of lungs by gamma/X-ray. In this direction, a few clinical trials have also been initiated. However, a few recent publications have raised some concerns regarding LDRT, especially about possibilities of activation/aggressiveness of virus (severe acute respiratory syndrome coronavirus 2 in case of COVID-19), lung injury and risk of second cancer after low-dose therapy. The present manuscript is an attempt to analyze these apprehensions based on cited references and other available literature, including some from our laboratory. At this point, LDRT may be not the first line of therapy. However, based on existing anti-inflammatory evidence of LDRT, it needs encouragement as an adjuvant therapy and for more multi-centric clinical trials. In addition, it would be worth combining LDRT with other anti-inflammatory therapies, which would open avenues for multi-modal therapy of pneumonia/ARDS in COVID-19 patients. The mode of irradiation (local lung irradiation or whole-body irradiation) and the window period after infection of the virus, need to be optimized using suitable animal studies for effective clinical outcomes of LDRT. However, considering ample evidence, it is time to look beyond the apprehensions if a low dose of radiation could be exploited for better management of COVID-19 patients.
Collapse
Affiliation(s)
- Badri Narain Pandey
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, Maharashtra, India
| |
Collapse
|
20
|
Radiotherapy-Mediated Immunomodulation and Anti-Tumor Abscopal Effect Combining Immune Checkpoint Blockade. Cancers (Basel) 2020; 12:cancers12102762. [PMID: 32992835 PMCID: PMC7600068 DOI: 10.3390/cancers12102762] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/19/2020] [Accepted: 09/23/2020] [Indexed: 12/17/2022] Open
Abstract
Radiotherapy (RT) is a conventional method for clinical treatment of local tumors, which can induce tumor-specific immune response and cause the shrinkage of primary tumor and distal metastases via mediating tumor infiltration of CD8+ T cells. Ionizing radiation (IR) induced tumor regression outside the radiation field is termed as abscopal effect. However, due to the mobilization of immunosuppressive signals by IR, the activated CD8+T cells are not sufficient to maintain a long-term positive feedback to make the tumors regress completely. Eventually, the "hot" tumors gradually turn to "cold". With the advent of emerging immunotherapy, the combination of immune checkpoint blockade (ICB) and local RT has produced welcome changes in stubborn metastases, especially anti-PD-1/PD-L1 and anti-CTLA-4 which have been approved in clinical cancer treatment. However, the detailed mechanism of the abscopal effect induced by combined therapy is still unclear. Therefore, how to formulate a therapeutic schedule to maximize the efficacy should be took into consideration according to specific circumstance. This paper reviewed the recent research progresses in immunomodulatory effects of local radiotherapy on the tumor microenvironment, as well as the unique advantage for abscopal effect when combined with ICB, with a view to exploring the potential application value of radioimmunotherapy in clinic.
Collapse
|
21
|
Cuiju W, Shibiao S, Ying T, Rongzong L, Haijuan X, Huifeng C, Tianjian W. IL-2 and IL-2R gene polymorphisms and immune function in people residing in areas with high background radiation, Yangjiang, China. Int J Radiat Biol 2020; 96:1466-1472. [PMID: 32910717 DOI: 10.1080/09553002.2020.1820607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE Long-term exposure to low dose radiation may trigger immune response and stimulate hormesis. Interleukin-2 (IL-2) and interleukin-2 receptor (IL-2R) play a crucial role in immune function. We aimed to explore the possible association of IL-2 and IL-2R gene polymorphisms with low dose radiation exposure, as well as the relationship with IL-2 gene expression in people residing in areas with a high background radiation in Yangjiang, China. MATERIALS AND METHODS We recruited and assigned 54 native men residing in Yangxi County, Yangjiang city to the high natural background radiation (HNBR) group, and 53 native men residing in Hengpi County, Enping city to the control area (CA) group. All the participants wore a thermoluminescent dosimeter (TLD) for 90 days, and answered questionnaires. The serum levels of IL2, IL4, IL5, sIL2R, and tumor growth factor (TGF), and expression levels of IL2RA, IL2RB, IL2RG, and IL2 were also analyzed. Additionally, we tested 10 polymorphic loci associated with the IL-2 gene. RESULTS The annual effective radiation doses in the HNBR and CA groups were 6.24 mSv y-1 and 1.95 mSv y-1, respectively. After adjusting for potential confounding factors, the serum levels of IL-2 and IL-5 were higher in the HNBR group than the CA group (p < .05), while the serum level of TGFβ was lower in the HNBR group (p < .05). The IL-2 gene mRNA expression level was higher in the HNBR group than the CA group (p < .05). The IL-2RB rs76206423 AA allele showed significant variations in the HNBR group (p = .0381). CONCLUSIONS Long-term exposure to low dose radiation may enhance immune function, and IL-2RB rs76206423 may be related to the expression of IL-2 by other coding variants. Moreover, our data provide a better understanding of the molecular mechanism of the immune response to low dose radiation.
Collapse
Affiliation(s)
- Wen Cuiju
- Guangdong Province Hospital for Occupational Disease Prevention and Treatment, Guangdong, China
| | - Su Shibiao
- Guangdong Province Hospital for Occupational Disease Prevention and Treatment, Guangdong, China
| | - Tang Ying
- Guangdong Province Hospital for Occupational Disease Prevention and Treatment, Guangdong, China
| | - Li Rongzong
- Guangdong Province Hospital for Occupational Disease Prevention and Treatment, Guangdong, China
| | - Xu Haijuan
- Guangdong Province Hospital for Occupational Disease Prevention and Treatment, Guangdong, China
| | - Chen Huifeng
- Guangdong Province Hospital for Occupational Disease Prevention and Treatment, Guangdong, China
| | - Wang Tianjian
- Guangdong Province Hospital for Occupational Disease Prevention and Treatment, Guangdong, China
| |
Collapse
|
22
|
Koca G, Singar E, Akbulut A, Yazihan N, Yumuşak N, Demir A, Korkmaz M. The Effect of Resveratrol on Radioiodine Therapy-Associated Lacrimal Gland Damage. Curr Eye Res 2020; 46:398-407. [PMID: 32730712 DOI: 10.1080/02713683.2020.1803920] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE We have evaluated the potential radioprotective, antioxidant and anti-apoptotic effects of resveratrol (RSV) against high-dose radioactive iodine (RAI) therapy associated damage of the lacrimal glands by biochemical, histopathological and immunohistochemical methods. MATERIALS AND METHODS Thirty Wistar-albino rats were randomly divided into three groups; the control group received no treatment or medication, the RAI group received RAI but no medication and the RSV group received oral RAI and intraperitoneal RSV. RSV was started at day one, before RAI administration, and continued for 8 days. Bilateral intraorbital (IG), extraorbital (EG), and Harderian (HG) lacrimal glands were evaluated in all rats for histopathological, immunohistochemical, tissue cytokine and oxidant and antioxidant level assessment. RESULTS RSV group restored inflammation, fibrosis, vacuolization, change in nucleus characteristics, lipofuscin-like accumulation and cellular morphologic patterns were statistically significant in all lacrimal gland types, compared to the RAI group (p < .05 for all variables). Similarly, elevated Caspase-3 and TUNEL levels in the RAI group were significantly alleviated in the RSV group in all lacrimal gland types (p < .05 for all variables). RAI administration significantly elevated TNF-α, IL-6, NF-кb levels, and decreased IL-10 levels (p < .05 for all parameters) whereas TOS levels significantly increased and TAS levels were significantly decreased. However, RSV significantly diminished TNF-α, IL-6, IL-4, and NF-кb levels. Furthermore, RSV significantly decreased TOS and increased TAS levels (p < .05 for all variables). CONCLUSIONS We conclude that with its anti-cancer effect as well as its antioxidant effect RSV has protected the histopathological pattern of the lacrimal glands from the damage, decreased inflammation in histopathologic assessments, and decreased tissue cytokine levels, apoptosis and DNA fragmentation on the lacrimal glands after RAI.
Collapse
Affiliation(s)
- Gökhan Koca
- Department of Nuclear Medicine, Ankara Training and Application Hospital, University of Health Sciences , Ankara, Turkey
| | - Evin Singar
- Department of Ophthalmology, Ankara Training and Application Hospital, University of Health Sciences , Ankara, Turkey
| | - Aylin Akbulut
- Department of Nuclear Medicine, Ankara Training and Application Hospital, University of Health Sciences , Ankara, Turkey
| | - Nuray Yazihan
- Department of Pathophysiology, Ankara University School of Medicine , Ankara, Turkey
| | - Nihat Yumuşak
- Department of Pathology, Harran University Faculty of Veterinary Medicine , Sanliurfa, Turkey
| | - Ayten Demir
- Faculty of Health Sciences, Nursing Department, Ankara University , Ankara, Turkey
| | - Meliha Korkmaz
- Department of Nuclear Medicine, Ankara Training and Application Hospital, University of Health Sciences , Ankara, Turkey
| |
Collapse
|
23
|
Farid A, Kamel D, Abdelwahab Montaser S, Mohamed Ahmed M, El Amir M, El Amir A. Synergetic role of senna and fennel extracts as antioxidant, anti-inflammatory and anti-mutagenic agents in irradiated human blood lymphocyte cultures. JOURNAL OF RADIATION RESEARCH AND APPLIED SCIENCES 2020. [DOI: 10.1080/16878507.2020.1723948] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Alyaa Farid
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Dina Kamel
- Radiation Protection Department, Nuclear Power Plant Authority (NPPA), Nasr City, Cairo, Egypt
| | - Sherien Abdelwahab Montaser
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Nasr City, Cairo, Egypt
| | - Mahmoud Mohamed Ahmed
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Nasr City, Cairo, Egypt
| | - Mona El Amir
- Internal Medicine Department, Faculty of Medicine, Kasr Al-Ainy, Cairo University, Giza, Egypt
| | - Azza El Amir
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
24
|
Ito T, Yamamoto T. Wells syndrome after radiation therapy. J Dermatol 2020; 47:e75-e77. [PMID: 31960465 DOI: 10.1111/1346-8138.15217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Takashi Ito
- Department of Dermatology, Fukushima Medical University, Fukushima, Japan
| | - Toshiyuki Yamamoto
- Department of Dermatology, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
25
|
Alosaimi B, Hamed ME, Naeem A, Alsharef AA, AlQahtani SY, AlDosari KM, Alamri AA, Al-Eisa K, Khojah T, Assiri AM, Enani MA. MERS-CoV infection is associated with downregulation of genes encoding Th1 and Th2 cytokines/chemokines and elevated inflammatory innate immune response in the lower respiratory tract. Cytokine 2019; 126:154895. [PMID: 31706200 PMCID: PMC7128721 DOI: 10.1016/j.cyto.2019.154895] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/14/2019] [Accepted: 10/17/2019] [Indexed: 12/31/2022]
Abstract
MERS-CoV infection downregulates Th1 and Th2 cytokines and chemokines. MERS-CoV infection provokes high levels of IL-1α, IL-1β and IL-8 (CXCL8). Inflammatory cytokines/chemokines correlate with MERS-CoV case fatality rate. Th1/Th2 downregulation may contribute to severe infection and evolution of ARDS.
MERS-CoV, a highly pathogenic virus in humans, is associated with high morbidity and case fatality. Inflammatory responses have a significant impact on MERS-CoV pathogenesis and disease outcome. However, CD4+ T-cell induced immune responses during acute MERS-CoV infection are barely detectable, with potent inhibition of effector T cells and downregulation of antigen presentation. The local pulmonary immune response, particularly the Th1 and Th2-related immune response during acute severe MERS-CoV infection is not fully understood. In this study, we offer the first insights into the pulmonary gene expression profile of Th1 and Th2-related cytokines/chemokines (Th1 & Th2 responses) during acute MERS-CoV infection using RT2 Profiler PCR Arrays. We also quantified the expression level of primary inflammatory cytokines/chemokines. Our results showed a downregulation of Th2, inadequate (partial) Th1 immune response and high expression levels of inflammatory cytokines IL-1α and IL-1β and the neutrophil chemoattractant chemokine IL-8 (CXCL8) in the lower respiratory tract of MERS-CoV infected patients. Moreover, we identified a high viral load in all included patients. We also observed a correlation between inflammatory cytokines, Th1, and Th2 downregulation and the case fatality rate. Th1 and Th2 response downregulation, high expression of inflammatory cytokines, and high viral load may contribute to lung inflammation, severe infection, the evolution of pneumonia and ARDS, and a higher case fatality rate. Further study of the molecular mechanisms underlying the Th1 and Th2 regulatory pathways will be vital for active vaccine development and the identification of novel therapeutic strategies.
Collapse
Affiliation(s)
- Bandar Alosaimi
- Research Center, King Fahad Medical City, Riyadh, Saudi Arabia; College of Medicine, King Fahad Medical City, Riyadh, Saudi Arabia.
| | - Maaweya E Hamed
- College of Science, King Saud University, Department of Botany and Microbiology, Riyadh, Saudi Arabia
| | - Asif Naeem
- Research Center, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Ali A Alsharef
- General Directorate of Laboratories and Blood Banks, Ministry of Health, Saudi Arabia
| | - Saeed Y AlQahtani
- General Directorate of Laboratories and Blood Banks, Ministry of Health, Saudi Arabia
| | - Kamel M AlDosari
- Riyadh Regional Laboratory, Ministry of Health, Riyadh, Saudi Arabia
| | - Aref A Alamri
- Riyadh Regional Laboratory, Ministry of Health, Riyadh, Saudi Arabia
| | - Kholoud Al-Eisa
- Riyadh Regional Laboratory, Ministry of Health, Riyadh, Saudi Arabia
| | - Taghreed Khojah
- Riyadh Regional Laboratory, Ministry of Health, Riyadh, Saudi Arabia
| | - Abdullah M Assiri
- Preventive Medicine Assistant Deputyship, Ministry of Health, Riyadh, Saudi Arabia
| | - Mushira A Enani
- Medical Specialties Department, Section of Infectious Diseases, King Fahad Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
26
|
Jang H, Lee J, Park S, Kim JS, Shim S, Lee SB, Han SH, Myung H, Kim H, Jang WS, Lee SJ, Myung JK. Baicalein Mitigates Radiation-Induced Enteritis by Improving Endothelial Dysfunction. Front Pharmacol 2019; 10:892. [PMID: 31474856 PMCID: PMC6707809 DOI: 10.3389/fphar.2019.00892] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 07/16/2019] [Indexed: 12/16/2022] Open
Abstract
Background and Aims: Radiation-induced intestinal injury occurred in application of radiotherapy for abdominal and pelvic cancers or in nuclear accidents. Radiation-induced enteritis may be considered an ideal model of gastrointestinal inflammation. The endothelium is a crucial component of inflammation, and the endothelial dysfunction following radiation exposure induces the intestinal proinflammatory response and progression of radiation enteritis. Baicalein (5,6,7-trihydroxyflavonoid) is a flavonoid from Scutellaria baicalensis used in oriental herbal medicine. Baicalein has been found to have multiple beneficial properties including antioxidant, anti-inflammatory, anti-allergic, and anti-cancer activities. Here, we investigated the therapeutic effects of baicalein on endothelial dysfunction in radiation-induced intestinal inflammation. Materials and Methods: We performed histological analysis, bacterial translocation, and intestinal permeability assays and also assessed infiltration of leukocytes and inflammatory cytokine expression using a mouse model of radiation-induced enteritis. In addition, to investigate the effect of baicalein in endothelial dysfunction, we analyzed endothelial-derived adherent molecules in human umbilical vein endothelial cells (HUVECs) and irradiated intestinal tissue. Results: Histological damage such as shortening of villi length and impaired intestinal crypt function was observed in the radiation-induced enteritis mouse model. Intestinal damage was attenuated in baicalein-treated groups with improvement of intestinal barrier function. Baicalein inhibited the expression of radiation-induced adherent molecules in HUVECs and intestine of irradiated mouse and decreased leukocyte infiltration in the radiation-induced enteritis. Conclusions: Baicalein could accelerate crypt regeneration via recovery of endothelial damage. Therefore, baicalein has a therapeutic effect on radiation-induced intestinal inflammation by attenuating endothelial damage.
Collapse
Affiliation(s)
- Hyosun Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Janet Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Sunhoo Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea.,Department of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Joong Sun Kim
- Herbal Medicine Resources Center, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Sehwan Shim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Seung Bum Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Sung-Honn Han
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Hyunwook Myung
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Hyewon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Won-Suk Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Sun-Joo Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Jae Kyung Myung
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea.,Department of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| |
Collapse
|
27
|
Hatiboglu MA, Kocyigit A, Guler EM, Nalli A, Akdur K, Sakarcan A, Ozek E, Uysal O, Mayadagli A. Gamma knife radiosurgery compared to whole brain radiation therapy enhances immunity via immunoregulatory molecules in patients with metastatic brain tumours. Br J Neurosurg 2019; 34:604-610. [PMID: 31317782 DOI: 10.1080/02688697.2019.1642445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Background: There is lack of data on the effect of stereotactic radiosurgery in modulation of the immune system for cancer patients with metastatic brain tumours. Therefore, we investigated the change in levels of immunoregulatory molecules after Gamma Knife radiosurgery (GKR) and whole brain radiation therapy (WBRT) in patients with brain metastases.Methods: Peripheral blood samples were collected from 15 patients who received GKR, nine patients who received WBRT for brain metastases and 10 healthy controls. Samples were obtained at three time points such as before, 1h after and 1 week after the index procedure for patients treated with GKR or WBRT. All patients' demographic data and radiosurgical parameters were retrospectively reviewed. We analyzed the change in the levels of T-lymphocyte-associated antigen 4 (CTLA-4) and programmed cell death ligand-1 (PD-L1), and cytokines such as IL-2, IL-10, IFN-γ, TNF-α after GKR and WBRT using Enzyme-linked immunosorbent assays (ELISA).Results: Baseline level of IFN-γ was found to be lower and that of PD-L1 was higher in the GKR group compared to WBRT group and healthy controls (p < 0.05 and p < 0.01, respectively). Levels of IFN-γ and IL-2 were increased (p < 0.01 and p < 0.01, respectively), while CTLA-4 and PD-L1 were decreased (p = 0.05 and p = 0.01, respectively) after GKR compared to pre-GKR levels, while there was no change after WBRT.Conclusion: GKR regulates immunoregulatory molecules towards enhancing the immune system, while WBRT did not exert any effect. These findings suggested that treatment of metastatic brain lesion with GKR might stimulate a systemic immune response against the tumour.
Collapse
Affiliation(s)
- Mustafa Aziz Hatiboglu
- Department of Neurosurgery, Bezmialem Vakif University School of Medicine, Istanbul, Turkey.,Department of Molecular Biology, Bezmialem Vakif University Beykoz Institute of Life Science and Biotechnology, Istanbul, Turkey
| | - Abdurrahim Kocyigit
- Department of Medical Biochemistry, Bezmialem Vakif University School of Medicine, Istanbul, Turkey
| | - Eray Metin Guler
- Department of Medical Biochemistry, Bezmialem Vakif University School of Medicine, Istanbul, Turkey
| | - Arife Nalli
- Department of Molecular Biology, Bezmialem Vakif University Beykoz Institute of Life Science and Biotechnology, Istanbul, Turkey
| | - Kerime Akdur
- Department of Neurosurgery, Bezmialem Vakif University School of Medicine, Istanbul, Turkey
| | - Ayten Sakarcan
- Department of Neurosurgery, Bezmialem Vakif University School of Medicine, Istanbul, Turkey
| | - Erdinc Ozek
- Department of Neurosurgery, Bezmialem Vakif University School of Medicine, Istanbul, Turkey
| | - Omer Uysal
- Department of Biostatistics, Bezmialem Vakif University School of Medicine, Istanbul, Turkey
| | - Alpaslan Mayadagli
- Department of Radiation Oncology, Bezmialem Vakif University School of Medicine, Istanbul, Turkey
| |
Collapse
|
28
|
Chen HY, Xie HY, Liu XX, Li LF, Bai YR, Gao JX. Splenic irradiation combined with tumor irradiation promotes T cell infiltration in the tumor microenvironment and helps in tumor control. Biochem Biophys Res Commun 2019; 510:156-162. [DOI: 10.1016/j.bbrc.2019.01.071] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 01/15/2019] [Indexed: 12/25/2022]
|
29
|
Dalgleish AG, Stern PL. The failure of radical treatments to cure cancer: can less deliver more? Ther Adv Vaccines Immunother 2018; 6:69-76. [PMID: 30623172 PMCID: PMC6304701 DOI: 10.1177/2515135518815393] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 11/05/2018] [Indexed: 12/31/2022] Open
Abstract
All too often attempts to deliver improved cancer cure rates by increasing the dose of a particular treatment are not successful enough to justify the accompanying increase in toxicity and reduction in quality of life suffered by a significant number of patients. In part, this drive for using higher levels of treatment derives from the nature of the process for testing and incorporation of new protocols. Indeed, new treatment regimens must now consider the key role of immunity in cancer control, a component that has been largely ignored until very recently. The recognition that some drugs developed for cytotoxicity at higher doses can display alternative anticancer activities at lower doses including through modulation of immune responses is prompting a significant re-evaluation of treatment protocol development. Given that tumours are remarkably heterogeneous and with inherent genetic instability it is probably only the adaptive immune response with its flexibility and extensive repertoire that can rise to the challenge of effecting significant control and ultimately elimination of a patient's cancer. This article discusses some of the elements that have limited higher levels of treatment outcomes and where too much proved less effective. We explore observations that less can often be as effective, if not more effective especially with some chemotherapy regimens, and discuss how this can be exploited in combination with immunotherapies to deliver nontoxic improved tumour responses.
Collapse
Affiliation(s)
- Angus G Dalgleish
- Infection and Immunity Centre, St George's, University of London, Cranmer Terrace, London, UK
| | - Peter L Stern
- Division of Molecular and Clinical Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Cancer Research Centre, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| |
Collapse
|