1
|
Ariyoshi K, Imaoka T, Ohmachi Y, Ishida Y, Uda M, Nishimura M, Shinagawa M, Yoshida M, Ogiu T, Kaminishi M, Morioka T, Kakinuma S, Shimada Y. Influence of Age on Leukemia Mortality Associated with Exposure to γ rays and 2-MeV Fast Neutrons in Male C3H Mice. Radiat Res 2024; 202:685-696. [PMID: 39187269 DOI: 10.1667/rade-23-00069.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/23/2024] [Indexed: 08/28/2024]
Abstract
The relative biological effectiveness (RBE) of densely ionizing radiation can depend on the biological context. From a radiological perspective, age is an important factor affecting health risks of radiation exposure, but little is known about the modifying impact of age on the effects of densely ionizing radiation. Herein, we addressed the influence of age on leukemogenesis induced by accelerator-generated fast neutrons (mean energy, ∼2 MeV). Male C3H/HeNrs mice were exposed to 137Cs γ rays (0.2-3.0 Gy) or neutrons (0.0485-0.97 Gy, γ ray contamination 0.0105-0.21 Gy) at 1, 3, 8, or 35 weeks of age and observed over their lifetimes under specific pathogen-free conditions. Leukemia and lymphoma were diagnosed pathologically. Hazard ratio (HR) and RBE for myeloid leukemia mortality as well as the age dependence of these two parameters were modeled and analyzed using Cox regression. Neutron exposure increased HR concordant with a linear dose response. The increase of HR per dose depended on age at exposure, with no significant dose dependence at age 1 or 3 weeks but a significant increase in HR of 5.5 per Gy (γ rays) and 16 per Gy (neutrons) at 8 weeks and 5.8 per Gy (γ rays) and 9 per Gy (neutrons) at 35 weeks. The RBE of neutrons was 2.1 (95% confidence interval, 1.1-3.7), with no dependence on age. The development of lymphoid neoplasms was not related to radiation exposure. The observed increasing trend of radiation-associated mortality of myeloid leukemia with age at exposure supports previous epidemiological and experimental findings. The results also suggest that exposure at the susceptible age of 8 or 35 weeks does not significantly influence the RBE value for neutrons for induction of leukemia, unlike what has been documented for breast and brain tumors.
Collapse
Affiliation(s)
- Kentaro Ariyoshi
- Research Center for Radiation Protection, National Institute of Radiological Sciences, Chiba, Japan
| | - Tatsuhiko Imaoka
- Department of Radiation Effects Research, Institute for Radiological Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Yasushi Ohmachi
- Research Center for Radiation Protection, National Institute of Radiological Sciences, Chiba, Japan
| | - Yuka Ishida
- Laboratory Animal and Genome Sciences Section, Department of Safety Administration, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Masahiro Uda
- Research Center for Radiation Protection, National Institute of Radiological Sciences, Chiba, Japan
| | - Mayumi Nishimura
- Department of Radiation Effects Research, Institute for Radiological Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Mayumi Shinagawa
- Department of Radiation Effects Research, Institute for Radiological Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Midori Yoshida
- Research Center for Radiation Protection, National Institute of Radiological Sciences, Chiba, Japan
| | - Toshiaki Ogiu
- Research Center for Radiation Protection, National Institute of Radiological Sciences, Chiba, Japan
| | - Mutsumi Kaminishi
- Department of Radiation Effects Research, Institute for Radiological Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Takamitsu Morioka
- Department of Radiation Effects Research, Institute for Radiological Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Shizuko Kakinuma
- Department of Radiation Effects Research, Institute for Radiological Science, National Institutes for Quantum Science and Technology, Chiba, Japan
| | - Yoshiya Shimada
- Department of Radiation Effects Research, Institute for Radiological Science, National Institutes for Quantum Science and Technology, Chiba, Japan
- Institute for Environmental Sciences, Kamikita-gun, Aomori, Japan
| |
Collapse
|
2
|
Zhou G, Shimura T, Yoneima T, Nagamachi A, Kanai A, Doi K, Sasatani M. Age-Dependent Differences in Radiation-Induced DNA Damage Responses in Intestinal Stem Cells. Int J Mol Sci 2024; 25:10213. [PMID: 39337697 PMCID: PMC11431935 DOI: 10.3390/ijms251810213] [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: 08/28/2024] [Revised: 09/20/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
Age at exposure is a critical modifier of the risk of radiation-induced cancer. However, the effects of age on radiation-induced carcinogenesis remain poorly understood. In this study, we focused on tissue stem cells using Lgr5-eGFP-ires-CreERT2 mice to compare radiation-induced DNA damage responses between Lgr5+ and Lgr5- intestinal stem cells. Three-dimensional immunostaining analyses demonstrated that radiation induced apoptosis and the mitotic index more efficiently in adult Lgr5- stem cells than in adult Lgr5+ stem cells but not in infants, regardless of Lgr5 expression. Supporting this evidence, rapid and transient p53 activation occurred after irradiation in adult intestinal crypts but not in infants. RNA sequencing revealed greater variability in gene expression in adult Lgr5+ stem cells than in infant Lgr5+ stem cells after irradiation. Notably, the cell cycle and DNA repair pathways were more enriched in adult stem cells than in infant stem cells after irradiation. Our findings suggest that radiation-induced DNA damage responses in mouse intestinal crypts differ between infants and adults, potentially contributing to the age-dependent susceptibility to radiation carcinogenesis.
Collapse
Grants
- none Research project on the Health Effects of Radiation organized by Ministry of the Environment, Japan.
- 23K25008 Japan Society for the Promotion of Science, JSPS KAKENHI
- 22H03754 Japan Society for the Promotion of Science, JSPS KAKENHI
- 23K28232 Japan Society for the Promotion of Science, JSPS KAKENHI
- 23H03542 Japan Society for the Promotion of Science, JSPS KAKENHI
- 20K21846 Japan Society for the Promotion of Science, JSPS KAKENHI
- NIFS20KOCA004 National Institute for Fusion Science Collaborative Research Program
- NIFS23HDCF005 National Institute for Fusion Science Collaborative Research Program
- none QST Research Collaboration
- none the Program of the Network-Type Joint Usage/Research Center for Radiation Disaster Medical Science at Hiroshima University, Nagasaki University, and Fukushima Medical University.
- none Initiative for Realizing Diversity in the Research Environment (Specific Correspondence Type), a support project for the Development of Human Resources in Science and Technology conducted by the Ministry of Education, Culture, Sports, Science and Technolo
- NIFS17KOCA002 National Institute for Fusion Science Collaborative Research Program
Collapse
Affiliation(s)
- Guanyu Zhou
- Department of Experimental Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 754-8553, Japan;
| | - Tsutomu Shimura
- Department of Environmental Health, National Institute of Public Health, Saitama 351-0197, Japan
| | - Taiki Yoneima
- School of Medicine, Hiroshima University, Hiroshima 754-8551, Japan
| | - Akiko Nagamachi
- Department of Molecular Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 754-8553, Japan
| | - Akinori Kanai
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba 277-8561, Japan
| | - Kazutaka Doi
- Department of Radiation Regulatory Science Research, Institute for Radiological Sciences, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Megumi Sasatani
- Department of Experimental Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 754-8553, Japan;
| |
Collapse
|
3
|
Koga F. Selective multimodal bladder-sparing therapy for muscle-invasive bladder cancer: the present and the future. Expert Rev Anticancer Ther 2023; 23:1127-1139. [PMID: 37753554 DOI: 10.1080/14737140.2023.2257389] [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: 07/10/2023] [Accepted: 09/06/2023] [Indexed: 09/28/2023]
Abstract
INTRODUCTION Despite the lack of level 1 evidence, selective bladder-sparing therapy using trimodal therapy is currently recommended by guidelines as a standard of care in patients with non-metastatic, muscle-invasive bladder cancer who are eligible for the treatment. AREAS COVERED This article reviews major studies of selective, bladder-sparing therapy utilizing multiple modalities for muscle-invasive bladder cancer and those comparing the oncological outcomes between bladder-sparing therapy and radical cystectomy. Also discussed are predictive biomarkers potentially capable of guiding treatment decisions by patients with muscle-invasive bladder cancer and a novel strategy for boosting the antitumor immune response in bladder-sparing therapy. PubMed databases were searched for records of 30 June 2023 or earlier. EXPERT OPINION Selective, bladder-sparing therapy appears to be underutilized at present. To promote its use, measures should be taken to facilitate the referral of eligible patients to specialist centers and broaden the number of facilities providing the therapy. Recent studies have suggested a prognostic benefit of radiotherapy for the primary lesion in patients with metastatic bladder cancer. Given that irradiation can induce the abscopal effect, particularly in combination with immune checkpoint inhibitors, demand for bladder-sparing therapies may increase in the context of treatments for metastases.
Collapse
Affiliation(s)
- Fumitaka Koga
- Department of Urology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
4
|
Massive expansion of multiple clones in the mouse hematopoietic system long after whole-body X-irradiation. Sci Rep 2022; 12:17276. [PMID: 36241679 PMCID: PMC9568546 DOI: 10.1038/s41598-022-21621-6] [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: 07/29/2022] [Accepted: 09/29/2022] [Indexed: 01/06/2023] Open
Abstract
Clonal hematopoiesis (CH) is prevalent in the elderly and associates with hematologic malignancy and cardiovascular disease. Although the risk of developing these diseases increases with radiation doses in atomic-bomb survivors, the causal relationship between radiation exposure and CH is unclear. This study investigated whether radiation exposure induces CH in mice 12-18 months after 3-Gy whole-body irradiation. We found radiation-associated increases in peripheral blood myeloid cells and red blood cell distribution width (RDW). Deep sequencing of bone marrow and non-hematopoietic tissue cells revealed recurrent somatic mutations specifically in the hematopoietic system in 11 of 12 irradiated mice but none in 6 non-irradiated mice. The irradiated mice possessed mutations with variant allele frequencies (VAFs) of > 0.02 on an average of 5.8 per mouse; mutations with VAFs of > 0.1 and/or deletion were prevalent. Examining hematopoietic stem/progenitor cells in two irradiated mice revealed several mutations co-existing in the same clones and multiple independent clones that deliver 60-80% of bone marrow nuclear cells. Our results indicate development of massive CH due to radiation exposure. Moreover, we have characterized mutations in radiation-induced CH.
Collapse
|
5
|
Sunaoshi M, Blyth BJ, Shang Y, Tsuruoka C, Morioka T, Shinagawa M, Ogawa M, Shimada Y, Tachibana A, Iizuka D, Kakinuma S. Post-Irradiation Thymic Regeneration in B6C3F1 Mice Is Age Dependent and Modulated by Activation of the PI3K-AKT-mTOR Pathway. BIOLOGY 2022; 11:biology11030449. [PMID: 35336821 PMCID: PMC8945464 DOI: 10.3390/biology11030449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 11/23/2022]
Abstract
Simple Summary Because children have a long life expectancy relative to adults and their tissues and organs are growing and developing rapidly, the risk of radiation carcinogenesis for children is considered higher than that for adults. However, the underlying mechanism(s) is unclear. To uncover the mechanism, we previously revealed that principal causative genes in mouse thymic lymphomas arising in irradiated infants or adults as Pten or Ikzf1, respectively, suggesting that cells with mutation in these genes might be the origin of lymphomas arising after irradiation depending on age at exposure. Here, we clarified the age-dependent differences in thymus-cell dynamics in mice during the initial post-irradiation period. Our results demonstrate that the dynamics of thymocytes during the post-irradiation period depends on the age at exposure. For irradiated infants in particular, the number of proliferating cells increase dramatically, and this correlate with activation of the PI3K-AKT-mTOR pathway. Thus, we conclude that the PI3K-AKT-mTOR pathway in infants contributed, at least in part, to thymus-cell dynamics through the modification of cell proliferation and survival after irradiation, which may be associated with the risk of Pten mutation-associated thymic lymphoma. Abstract The risk of radiation-induced carcinogenesis depends on age at exposure. We previously reported principal causative genes in lymphomas arising after infant or adult exposure to 4-fractionated irradiation as Pten or Ikzf1, respectively, suggesting that cells with mutation in these genes might be the origin of lymphomas arising after irradiation depending on age at exposure. Here, we clarified the age-dependent differences in thymus-cell dynamics in mice during the initial post-irradiation period. The thymocyte number initially decreased, followed by two regeneration phases. During the first regeneration, the proportion of phosphorylated-AKT-positive (p-AKT+) cells in cell-cycle phases S+G2/M of immature CD4−CD8− and CD4+CD8+ thymocytes and in phases G0/G1 of mature CD4+CD8− and CD4−CD8+ thymocytes was significantly greater in irradiated infants than in irradiated adults. During the second regeneration, the proportion of p-AKT+ thymocytes in phases G0/G1 increased in each of the three populations other than CD4−CD8− thymocytes more so than during the first regeneration. Finally, PI3K-AKT-mTOR signaling in infants contributed, at least in part, to biphasic thymic regeneration through the modification of cell proliferation and survival after irradiation, which may be associated with the risk of Pten mutation-associated thymic lymphoma.
Collapse
Affiliation(s)
- Masaaki Sunaoshi
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Chiba 263-8555, Japan; (M.S.); (B.J.B.); (Y.S.); (C.T.); (T.M.); (M.S.); (M.O.); (Y.S.); (S.K.)
| | - Benjamin J. Blyth
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Chiba 263-8555, Japan; (M.S.); (B.J.B.); (Y.S.); (C.T.); (T.M.); (M.S.); (M.O.); (Y.S.); (S.K.)
| | - Yi Shang
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Chiba 263-8555, Japan; (M.S.); (B.J.B.); (Y.S.); (C.T.); (T.M.); (M.S.); (M.O.); (Y.S.); (S.K.)
| | - Chizuru Tsuruoka
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Chiba 263-8555, Japan; (M.S.); (B.J.B.); (Y.S.); (C.T.); (T.M.); (M.S.); (M.O.); (Y.S.); (S.K.)
| | - Takamitsu Morioka
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Chiba 263-8555, Japan; (M.S.); (B.J.B.); (Y.S.); (C.T.); (T.M.); (M.S.); (M.O.); (Y.S.); (S.K.)
| | - Mayumi Shinagawa
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Chiba 263-8555, Japan; (M.S.); (B.J.B.); (Y.S.); (C.T.); (T.M.); (M.S.); (M.O.); (Y.S.); (S.K.)
| | - Mari Ogawa
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Chiba 263-8555, Japan; (M.S.); (B.J.B.); (Y.S.); (C.T.); (T.M.); (M.S.); (M.O.); (Y.S.); (S.K.)
| | - Yoshiya Shimada
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Chiba 263-8555, Japan; (M.S.); (B.J.B.); (Y.S.); (C.T.); (T.M.); (M.S.); (M.O.); (Y.S.); (S.K.)
| | - Akira Tachibana
- Graduate School of Science and Engineering, Ibaraki University, 2-1-1 Bunkyo, Mito 310-8512, Japan;
| | - Daisuke Iizuka
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Chiba 263-8555, Japan; (M.S.); (B.J.B.); (Y.S.); (C.T.); (T.M.); (M.S.); (M.O.); (Y.S.); (S.K.)
- Correspondence: ; Tel.: +81-43-206-3160
| | - Shizuko Kakinuma
- Department of Radiation Effects Research, National Institute of Radiological Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Chiba 263-8555, Japan; (M.S.); (B.J.B.); (Y.S.); (C.T.); (T.M.); (M.S.); (M.O.); (Y.S.); (S.K.)
| |
Collapse
|
6
|
Sim HJ, So HS, Poudel SB, Bhattarai G, Cho ES, Lee JC, Kook SH. Osteoblastic Wls Ablation Protects Mice from Total Body Irradiation-Induced Impairments in Hematopoiesis and Bone Marrow Microenvironment. Aging Dis 2022; 14:919-936. [PMID: 37191410 DOI: 10.14336/ad.2022.1026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/26/2022] [Indexed: 11/18/2022] Open
Abstract
Ionizing irradiation (IR) causes bone marrow (BM) injury, with senescence and impaired self-renewal of hematopoietic stem cells (HSCs), and inhibiting Wnt signaling could enhance hematopoietic regeneration and survival against IR stress. However, the underlying mechanisms by which a Wnt signaling blockade modulates IR-mediated damage of BM HSCs and mesenchymal stem cells (MSCs) are not yet completely understood. We investigated the effects of osteoblastic Wntless (Wls) depletion on total body irradiation (TBI, 5 Gy)-induced impairments in hematopoietic development, MSC function, and the BM microenvironment using conditional Wls knockout mutant mice (Col-Cre;Wlsfl/fl) and their littermate controls (Wlsfl/fl). Osteoblastic Wls ablation itself did not dysregulate BM frequency or hematopoietic development at a young age. Exposure to TBI at 4 weeks of age induced severe oxidative stress and senescence in the BM HSCs of Wlsfl/fl mice but not in those of the Col-Cre;Wlsfl/fl mice. TBI-exposed Wlsfl/fl mice exhibited greater impairments in hematopoietic development, colony formation, and long-term repopulation than TBI-exposed Col-Cre;Wlsfl/fl mice. Transplantation with BM HSCs or whole BM cells derived from the mutant, but not Wlsfl/fl mice, protected against HSC senescence and hematopoietic skewing toward myeloid cells and enhanced survival in recipients of lethal TBI (10 Gy). Unlike the Wlsfl/fl mice, the Col-Cre;Wlsfl/fl mice also showed radioprotection against TBI-mediated MSC senescence, bone mass loss, and delayed body growth. Our results indicate that osteoblastic Wls ablation renders BM-conserved stem cells resistant to TBI-mediated oxidative injuries. Overall, our findings show that inhibiting osteoblastic Wnt signaling promotes hematopoietic radioprotection and regeneration.
Collapse
|
7
|
Nava S, Lisini D, Frigerio S, Bersano A. Dendritic Cells and Cancer Immunotherapy: The Adjuvant Effect. Int J Mol Sci 2021; 22:ijms222212339. [PMID: 34830221 PMCID: PMC8620771 DOI: 10.3390/ijms222212339] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/10/2021] [Accepted: 11/12/2021] [Indexed: 01/01/2023] Open
Abstract
Dendritic cells (DCs) are immune specialized cells playing a critical role in promoting immune response against antigens, and may represent important targets for therapeutic interventions in cancer. DCs can be stimulated ex vivo with pro-inflammatory molecules and loaded with tumor-specific antigen(s). Protocols describing the specific details of DCs vaccination manufacturing vary widely, but regardless of the employed protocol, the DCs vaccination safety and its ability to induce antitumor responses is clearly established. Many years of studies have focused on the ability of DCs to provide overall survival benefits at least for a selection of cancer patients. Lessons learned from early trials lead to the hypothesis that, to improve the efficacy of DCs-based immunotherapy, this should be combined with other treatments. Thus, the vaccine’s ultimate role may lie in the combinatorial approaches of DCs-based immunotherapy with chemotherapy and radiotherapy, more than in monotherapy. In this review, we address some key questions regarding the integration of DCs vaccination with multimodality therapy approaches for cancer treatment paradigms.
Collapse
|
8
|
Ariyoshi K, Miura T, Kasai K, Goh VST, Fujishima Y, Nakata A, Takahashi A, Shimizu Y, Shinoda H, Yamashiro H, Seymour C, Mothersill C, Yoshida MA. Environmental radiation on large Japanese field mice in Fukushima reduced colony forming potential in hematopoietic progenitor cells without inducing genomic instability. Int J Radiat Biol 2020; 98:1147-1158. [PMID: 32791031 DOI: 10.1080/09553002.2020.1807643] [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 To study the environmental radiation effects of wild animals after the Fukushima Dai-ichi nuclear power plant accident, we assessed effects on hematopoietic progenitor cells (HPCs) in large Japanese field mice (Apodemus speciosus). MATERIALS AND METHODS A. speciosus were collected from three contaminated sites and control area. The air dose-rates at the control and contaminated areas were 0.96 ± 0.05 μGy/d (Hirosaki), 14.4 ± 2.4 μGy/d (Tanashio), 208.8 ± 31.2 μGy/d (Ide), 470.4 ± 93.6 μGy/d (Omaru), respectively. We investigated possible DNA damage and pro-inflammatory markers in the bone marrow (BM) cells. The colony-forming potential of BM cells was estimated by the number of HPC colony-forming cells. Radiation-induced genomic instability (RIGI) in HPCs was also analyzed by quantifying delayed DNA damage in CFU-GM clones. RESULTS Although no significant differences in DNA damage and inflammation markers in BM cells from control and contaminated areas, the number of HPC colonies exhibited an inverse correlation with air dose-rate. With regard to RIGI, no significant differences in DNA damage of CFU-GM clones between the mice from the control and the three contaminated areas. CONCLUSIONS Our study suggests that low dose-rate radiation of more than 200 Gy/d reduced HPCs, possibly eliminating genomically unstable HPCs.
Collapse
Affiliation(s)
- Kentaro Ariyoshi
- Integrated Center for Science and Humanities, Fukushima Medical University, Fukushima City, Japan
| | - Tomisato Miura
- Department of Radiation Biology, Institute of Radiation Emergency Medicine, Hirosaki University, Hirosaki, Japan
| | - Kosuke Kasai
- Department of Bioscience and Laboratory Medicine, Hirosaki University Graduate School of Health Sciences, Hirosaki, Japan
| | - Valerie Swee Ting Goh
- Department of Bioscience and Laboratory Medicine, Hirosaki University Graduate School of Health Sciences, Hirosaki, Japan
| | - Yohei Fujishima
- Department of Radiation Biology, Tohoku University School of Medicine, Sendai, Japan
| | - Akifumi Nakata
- Faculty of Pharmaceutical Sciences, Hokkaido University of Science, Sapporo, Japan
| | | | | | - Hisashi Shinoda
- Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Hideaki Yamashiro
- Graduate School of Science and Technology, Niigata University, Nishiku, Japan
| | - Colin Seymour
- Department of Biology, McMaster University, Hamilton, Canada
| | | | - Mitsuaki A Yoshida
- Department of Radiation Biology, Institute of Radiation Emergency Medicine, Hirosaki University, Hirosaki, Japan
| |
Collapse
|
9
|
Applegate KE, Rühm W, Wojcik A, Bourguignon M, Brenner A, Hamasaki K, Imai T, Imaizumi M, Imaoka T, Kakinuma S, Kamada T, Nishimura N, Okonogi N, Ozasa K, Rübe CE, Sadakane A, Sakata R, Shimada Y, Yoshida K, Bouffler S. Individual response of humans to ionising radiation: governing factors and importance for radiological protection. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2020; 59:185-209. [PMID: 32146555 DOI: 10.1007/s00411-020-00837-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 02/26/2020] [Indexed: 05/23/2023]
Abstract
Tissue reactions and stochastic effects after exposure to ionising radiation are variable between individuals but the factors and mechanisms governing individual responses are not well understood. Individual responses can be measured at different levels of biological organization and using different endpoints following varying doses of radiation, including: cancers, non-cancer diseases and mortality in the whole organism; normal tissue reactions after exposures; and, cellular endpoints such as chromosomal damage and molecular alterations. There is no doubt that many factors influence the responses of people to radiation to different degrees. In addition to the obvious general factors of radiation quality, dose, dose rate and the tissue (sub)volume irradiated, recognized and potential determining factors include age, sex, life style (e.g., smoking, diet, possibly body mass index), environmental factors, genetics and epigenetics, stochastic distribution of cellular events, and systemic comorbidities such as diabetes or viral infections. Genetic factors are commonly thought to be a substantial contributor to individual response to radiation. Apart from a small number of rare monogenic diseases such as ataxia telangiectasia, the inheritance of an abnormally responsive phenotype among a population of healthy individuals does not follow a classical Mendelian inheritance pattern. Rather it is considered to be a multi-factorial, complex trait.
Collapse
Affiliation(s)
| | - W Rühm
- Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Radiation Medicine, Neuherberg, Germany
| | - A Wojcik
- Centre for Radiation Protection Research, MBW Department, Stockholm University, Stockholm, Sweden
| | - M Bourguignon
- Department of Biophysics and Nuclear Medicine, University of Paris Saclay (UVSQ), Verseilles, France
| | - A Brenner
- Department of Epidemiology, Radiation Effects Research Foundation, Hiroshima, Japan
| | - K Hamasaki
- Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima, Japan
| | - T Imai
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Sciences and Technology, Chiba, Japan
| | - M Imaizumi
- Department of Nagasaki Clinical Studies, Radiation Effects Research Foundation, Nagasaki, Japan
| | - T Imaoka
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institute for Quantum and Radiological Science and Technology, Chiba, Japan
| | - S Kakinuma
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institute for Quantum and Radiological Science and Technology, Chiba, Japan
| | - T Kamada
- QST Hospital, National Institute of Radiological Sciences, National Institute for Quantum and Radiological Science and Technology, Chiba, Japan
| | - N Nishimura
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institute for Quantum and Radiological Science and Technology, Chiba, Japan
| | - N Okonogi
- QST Hospital, National Institute of Radiological Sciences, National Institute for Quantum and Radiological Science and Technology, Chiba, Japan
| | - K Ozasa
- Department of Epidemiology, Radiation Effects Research Foundation, Hiroshima, Japan
| | - C E Rübe
- Department of Radiation Oncology, Saarland University Medical Center, Homburg/Saar, Germany
| | - A Sadakane
- Department of Epidemiology, Radiation Effects Research Foundation, Hiroshima, Japan
| | - R Sakata
- Department of Epidemiology, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Y Shimada
- National Institute for Quantum and Radiological Science and Technology, Chiba, Japan
- Institute for Environmental Sciences, Aomori, Japan
| | - K Yoshida
- Immunology Laboratory, Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima, Japan
| | - S Bouffler
- Radiation Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Chilto, Didcot, UK
| |
Collapse
|
10
|
Shevtsov M, Sato H, Multhoff G, Shibata A. Novel Approaches to Improve the Efficacy of Immuno-Radiotherapy. Front Oncol 2019; 9:156. [PMID: 30941308 PMCID: PMC6433964 DOI: 10.3389/fonc.2019.00156] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 02/25/2019] [Indexed: 12/31/2022] Open
Abstract
Radiotherapy (RT) has been applied for decades as a treatment modality in the management of various types of cancer. Ionizing radiation induces tumor cell death, which in turn can either elicit protective anti-tumor immune responses or immunosuppression in the tumor micromilieu that contributes to local tumor recurrence. Immunosuppression is frequently accompanied by the attraction of immunosuppressive cells such as myeloid-derived suppressor cells (MDSCs), M2 tumor-associated macrophages (TAMs), T regulatory cells (Tregs), N2 neutrophils, and by the release of immunosuppressive cytokines (TGF-β, IL-10) and chemokines. Immune checkpoint pathways, particularly of the PD-1/PD-L1 axis, have been determined as key regulators of cancer immune escape. While IFN-dependent upregulation of PD-L1 has been extensively investigated, up-to-date studies indicated the importance of DNA damage signaling in the regulation of PD-L1 expression following RT. DNA damage dependent PD-L1 expression is upregulated by ATM/ATR/Chk1 kinase activities and cGAS/STING-dependent pathway, proving the role of DNA damage signaling in PD-L1 induced expression. Checkpoint blockade immunotherapies (i.e., application of anti-PD-1 and anti-PD-L1 antibodies) combined with RT were shown to significantly improve the objective response rates in therapy of various primary and metastatic malignancies. Further improvements in the therapeutic potential of RT are based on combinations of RT with other immunotherapeutic approaches including vaccines, cytokines and cytokine inducers, and an adoptive immune cell transfer (DCs, NK cells, T cells). In the current review we provide immunological rationale for a combination of RT with various immunotherapies as well as analysis of the emerging preclinical evidences for these therapies.
Collapse
Affiliation(s)
- Maxim Shevtsov
- Center for Translational Cancer Research, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany.,Institute of Cytology, Russian Academy of Sciences (RAS), St. Petersburg, Russia.,First Pavlov State Medical University of St. Petersburg, St. Petersburg, Russia.,Almazov National Medical Research Centre, Polenov Russian Scientific Research Institute of Neurosurgery, St. Petersburg, Russia
| | - Hiro Sato
- Department of Radiation Oncology, Graduate School of Medicine, Gunma University, Maebashi, Japan
| | - Gabriele Multhoff
- Center for Translational Cancer Research, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
| | - Atsushi Shibata
- Education and Research Support Center, Graduate School of Medicine, Gunma University, Maebashi, Japan
| |
Collapse
|
11
|
Haematological analysis of Japanese macaques (Macaca fuscata) in the area affected by the Fukushima Daiichi Nuclear Power Plant accident. Sci Rep 2018; 8:16748. [PMID: 30425289 PMCID: PMC6233195 DOI: 10.1038/s41598-018-35104-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 10/28/2018] [Indexed: 12/25/2022] Open
Abstract
Several populations of wild Japanese macaques (Macaca fuscata) inhabit the area around Fukushima Daiichi Nuclear Power Plant (FNPP). To measure and control the size of these populations, macaques are captured annually. Between May 2013 and December 2014, we performed a haematological analysis of Japanese macaques captured within a 40-km radius of FNPP, the location of a nuclear disaster two years post-accident. The dose-rate of radiocaesium was estimated using the ERICA Tool. The median internal dose-rate was 7.6 μGy/day (ranging from 1.8 to 219 μGy/day) and the external dose-rate was 13.9 μGy/day (ranging from 6.7 to 35.1 μGy/day). We performed multiple regression analyses to estimate the dose-rate effects on haematological values in peripheral blood and bone marrow. The white blood cell and platelet counts showed an inverse correlation with the internal dose-rate in mature macaques. Furthermore, the myeloid cell, megakaryocyte, and haematopoietic cell counts were inversely correlated and the occupancy of adipose tissue was positively correlated with internal dose-rate in femoral bone marrow of mature macaques. These relationships suggest that persistent whole body exposure to low-dose-rate radiation affects haematopoiesis in Japanese macaques.
Collapse
|
12
|
Ariyoshi K, Miura T, Kasai K, Akifumi N, Fujishima Y, Yoshida MA. Age Dependence of Radiation-Induced Genomic Instability in Mouse Hematopoietic Stem Cells. Radiat Res 2018; 190:623. [PMID: 30311845 DOI: 10.1667/rr15113.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Age at exposure is a critical factor that influences the risk of radiation-induced leukemia. Accumulating evidence suggests that ionizing radiation can induce genomic instability and promote leukemogenesis in hematopoietic stem cells (HSCs); however, the influence of age on this phenomenon has not been elucidated. In this study, infant (1-week-old) or adult (14-week-old) C3H/He mice received sham or 4 Gy whole-body irradiation, and bone marrow cells were transplanted to recipients at day 1 or 60 postirradiation. Twelve days after bone marrow transplant, we analyzed the radiation-induced genomic instability by scoring the frequency of DNA damage and micronucleus formation in colony-forming units-spleen (CFU-Ss). We observed significant increases in DNA damage and micronucleus formation in CFU-Ss of the 4 Gy irradiated adult cells transplanted at day 1 or 60 postirradiation. However, the frequency of DNA damage focus and micronucleus formation in CFU-Ss of 4 Gy irradiated infant cells transplanted at day 1 or 60 postirradiation was relatively decreased. Quantitative differences in the reactive oxygen species and cells expressing inducible nitric oxide synthase in CFU-Ss suggested that age-dependent radiation-induced genomic instability may result from chronic oxidative stress by pro-inflammatory states in HSC descendants after radiation exposure.
Collapse
Affiliation(s)
- Kentaro Ariyoshi
- a Department of Radiation Biology, Institute of Radiation Emergency Medicine, Hirosaki University, Hirosaki 036-8564, Japan
| | - Tomisato Miura
- b Department of Biomedical Sciences, Hirosaki University Graduate School of Health Sciences, Hirosaki 036-8564, Japan
| | - Kosuke Kasai
- b Department of Biomedical Sciences, Hirosaki University Graduate School of Health Sciences, Hirosaki 036-8564, Japan
| | - Nakata Akifumi
- c Department of Basic Pharmacy, Hokkaido Pharmaceutical University School of Pharmacy, Otaru, 047-0264, Japan
| | - Yohei Fujishima
- b Department of Biomedical Sciences, Hirosaki University Graduate School of Health Sciences, Hirosaki 036-8564, Japan
| | - Mitsuaki A Yoshida
- a Department of Radiation Biology, Institute of Radiation Emergency Medicine, Hirosaki University, Hirosaki 036-8564, Japan
| |
Collapse
|
13
|
Boulle G, Velut Y, Boni A, Lupo A, Alifano M, Trédaniel J, Giraud P, Damotte D. [Analysis of immune microenvironment by multiplex immunohistochemistry in locally advanced non-small cell lung cancer: Principles and perspectives]. Cancer Radiother 2018; 22:688-693. [PMID: 30131267 DOI: 10.1016/j.canrad.2018.07.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 07/04/2018] [Indexed: 12/14/2022]
Abstract
Recent therapeutic advances in non-small cell lung cancer allow a better understanding of the interactions between the tumour and its direct immune environment. The identification of new immune biomarkers integrating both cell subpopulations and their interactions is a real issue in oncology. New techniques of tissue analysis, particularly multiplex immunohistochemistry, consisting of a labelling of several antigens of interest by immunofluorescence on the same slide, provide a better understanding of the tumour environment. Integration of these modalities of analysis to the therapeutic decision is promising, because it allows an increased characterization of each tumour, particularly interesting with radiotherapy and immunotherapy. This article describes the potential of these assays in locally advanced non-small cell lung cancer.
Collapse
Affiliation(s)
- G Boulle
- Centre de recherche des Cordeliers, Institut national de la santé et de la recherche médicale (Inserm), UMRS 1138, 15, rue de l'École-de-Médecine, 75006 Paris, France
| | - Y Velut
- Centre de recherche des Cordeliers, Institut national de la santé et de la recherche médicale (Inserm), UMRS 1138, 15, rue de l'École-de-Médecine, 75006 Paris, France
| | - A Boni
- Centre de recherche des Cordeliers, Institut national de la santé et de la recherche médicale (Inserm), UMRS 1138, 15, rue de l'École-de-Médecine, 75006 Paris, France
| | - A Lupo
- Centre de recherche des Cordeliers, Institut national de la santé et de la recherche médicale (Inserm), UMRS 1138, 15, rue de l'École-de-Médecine, 75006 Paris, France; Service d'anatomopathologie, hôpital Cochin, 27, rue du Faubourg-Saint-Jacques, 75014 Paris, France
| | - M Alifano
- Service de chirurgie thoracique, hôpital Cochin, 27, rue du Faubourg-Saint-Jacques, 75014 Paris, France
| | - J Trédaniel
- Service d'oncologie thoracique, groupe hospitalier Paris Saint-Joseph, 185, rue Raymond-Losserand, 75014 Paris, France
| | - P Giraud
- Service d'oncologie radiothérapie, hôpital européen Georges-Pompidou, 20, rue Leblanc, 75015 Paris, France
| | - D Damotte
- Centre de recherche des Cordeliers, Institut national de la santé et de la recherche médicale (Inserm), UMRS 1138, 15, rue de l'École-de-Médecine, 75006 Paris, France; Service d'anatomopathologie, hôpital Cochin, 27, rue du Faubourg-Saint-Jacques, 75014 Paris, France.
| |
Collapse
|
14
|
In Vivo Irradiation of Mice Induces Activation of Dendritic Cells. Int J Mol Sci 2018; 19:ijms19082391. [PMID: 30110907 PMCID: PMC6121955 DOI: 10.3390/ijms19082391] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 08/09/2018] [Indexed: 11/17/2022] Open
Abstract
It is becoming clear that ionizing radiation positively influences certain immune parameters, which opens the possibility for combining radio- and immunotherapies in cancer treatment. The presence of functionally competent dendritic cells (DCs) is crucial in mounting a successful antitumor immune response. While it has been shown that DCs are relatively radioresistant, few and contradictory data are available on how ionizing radiation alters the functional integrity of these cells. Therefore, our objective was to investigate the effect of whole-body irradiation on the function of splenic DCs. C57Bl/6 mice were irradiated with 0.1, 0.25, and 2 Gy X-rays and changes in the phenotype of splenic DCs were compared to unirradiated controls. An increase was seen in DC surface markers influencing DC-T cell interactions. In vivo cytokine production was determined by direct intracellular cytokine staining. Irradiation with 2 Gy induced a 1.6-fold increase in IL-1α production, while the combination of irradiation and lipopolysaccharide (LPS) treatment induced a 3.9-fold increase, indicating a strong synergism between irradiation and LPS stimulation. Interaction of DCs with effector and regulatory T cells was investigated in a mixed lymphocyte reaction. While DCs from control animals induced stronger proliferation of regulatory T cells, DCs from animals irradiated with 2 Gy induced stronger proliferation of effector T cells. Antigen uptake and presentation was investigated by measuring the capacity of DCs to internalize and present ovalbumine (OVA)-derived peptides on their major histocompatibility complex (MHCI) molecules. Irradiation with 2 Gy did not influence antigen uptake or presentation, while low doses stimulated antigen uptake and reduced the level of antigen presentation. In conclusion, high-dose in vivo irradiation induced increased expression of T cell costimulatory markers, enhanced production of proinflammatory cytokines and a stronger stimulation of effector T cell proliferation than that of regulatory T cells. However, it did not influence DC antigen uptake or presentation. On the other hand, low-dose irradiation increased antigen uptake and lowered antigen presentation of DCs, indicating that low- and high-dose irradiation act on different pathways in DCs.
Collapse
|
15
|
Monzen S, Chiba M, Ueno T, Morino Y, Terada K, Yamaya H, Hosokawa Y. A radioresistant fraction of acute promyelocytic leukemia cells exhibit CD38 cell-surface antigen and mRNA expression. Oncol Lett 2018; 15:6709-6714. [PMID: 29616132 DOI: 10.3892/ol.2018.8099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 12/05/2017] [Indexed: 11/06/2022] Open
Abstract
In the present study, the cell viability and cluster of differentiation (CD)38 mRNA expression were evaluated in radioresistant (Res)-HL60 acute promyelocytic leukemia (APL) cells. Cell viability in Res-HL60 cells was higher compared with wild-type HL60 cells, but did not differ between high and mid/low CD38 antigen expression groups in Res-HL60 cells. A higher expression of CD38 mRNA in Res-HL60 cells was observed, particularly in the CD38high cell subpopulation. Furthermore, the expression of CD38 mRNA was upregulated following exposure to X-irradiation. In contrast, the characteristic expression of CD45 and CCAAT/enhancer-binding protein α mRNA were not altered. These results suggest that the accumulation of CD38 protein in radioresistant APL cells, resulting from the constant expression of CD38 mRNA induced by X-irradiation, is a characteristic response of the radioresistant-surviving fraction; however, the accumulation of CD38 did not influence the extent of radioresistant behavior.
Collapse
Affiliation(s)
- Satoru Monzen
- Department of Radiation Sciences, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori 036-8564, Japan
| | - Mitsuru Chiba
- Department of Bioscience and Laboratory Medicine, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori 036-8564, Japan
| | - Tatsuya Ueno
- Department of Radiation Sciences, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori 036-8564, Japan
| | - Yuki Morino
- Department of Radiation Sciences, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori 036-8564, Japan
| | - Kenji Terada
- Department of Radiation Sciences, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori 036-8564, Japan
| | - Hiroki Yamaya
- Department of Radiation Sciences, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori 036-8564, Japan
| | - Yoichiro Hosokawa
- Department of Radiation Sciences, Hirosaki University Graduate School of Health Sciences, Hirosaki, Aomori 036-8564, Japan
| |
Collapse
|
16
|
Szatmári T, Persa E, Kis E, Benedek A, Hargitai R, Sáfrány G, Lumniczky K. Extracellular vesicles mediate low dose ionizing radiation-induced immune and inflammatory responses in the blood. Int J Radiat Biol 2018. [PMID: 29533121 DOI: 10.1080/09553002.2018.1450533] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE Radiation-induced bystander effects (RIBE) imply the involvement of complex signaling mechanisms, which can be mediated by extracellular vesicles (EVs). Using an in vivo model, we investigated EV-transmitted RIBE in blood plasma and radiation effects on plasma EV miRNA profiles. MATERIALS AND METHODS C57Bl/6 mice were total-body irradiated with 0.1 and 2 Gy, bone marrow-derived EVs were isolated, and injected systemically into naive, 'bystander' animals. Proteome profiler antibody array membranes were used to detect alterations in plasma, both in directly irradiated and bystander mice. MiRNA profile of plasma EVs was determined by PCR array. RESULTS M-CSF and pentraxin-3 levels were increased in the blood of directly irradiated and bystander mice both after low and high dose irradiations, CXCL16 and lipocalin-2 increased after 2 Gy in directly irradiated and bystander mice, CCL5 and CCL11 changed in bystander mice only. Substantial overlap was found in the cellular pathways regulated by those miRNAs whose level were altered in EVs isolated from the plasma of mice irradiated with 0.1 and 2 Gy. Several of these pathways have already been associated with bystander responses. CONCLUSION Low and high dose effects overlapped both in EV-mediated alterations in signaling pathways leading to RIBE and in their systemic manifestations.
Collapse
Affiliation(s)
- Tünde Szatmári
- a Department of Radiation Medicine, Division of Radiobiology and Radiohygiene , National Public Health Institute , Budapest , Hungary
| | - Eszter Persa
- a Department of Radiation Medicine, Division of Radiobiology and Radiohygiene , National Public Health Institute , Budapest , Hungary
| | - Enikő Kis
- a Department of Radiation Medicine, Division of Radiobiology and Radiohygiene , National Public Health Institute , Budapest , Hungary
| | - Anett Benedek
- a Department of Radiation Medicine, Division of Radiobiology and Radiohygiene , National Public Health Institute , Budapest , Hungary
| | - Rita Hargitai
- a Department of Radiation Medicine, Division of Radiobiology and Radiohygiene , National Public Health Institute , Budapest , Hungary
| | - Géza Sáfrány
- a Department of Radiation Medicine, Division of Radiobiology and Radiohygiene , National Public Health Institute , Budapest , Hungary
| | - Katalin Lumniczky
- a Department of Radiation Medicine, Division of Radiobiology and Radiohygiene , National Public Health Institute , Budapest , Hungary
| |
Collapse
|
17
|
Shang Y, Sawa Y, Blyth BJ, Tsuruoka C, Nogawa H, Shimada Y, Kakinuma S. Radiation Exposure Enhances Hepatocyte Proliferation in Neonatal Mice but not in Adult Mice. Radiat Res 2017; 188:235-241. [DOI: 10.1667/rr14563.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Yi Shang
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Yurika Sawa
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Benjamin J. Blyth
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Chizuru Tsuruoka
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Hiroyuki Nogawa
- Department of Biology, Graduate School of Science, Chiba University, 1-33, Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan
| | - Yoshiya Shimada
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| | - Shizuko Kakinuma
- Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba 263-8555, Japan
| |
Collapse
|
18
|
Sharabi AB, Lim M, DeWeese TL, Drake CG. Radiation and checkpoint blockade immunotherapy: radiosensitisation and potential mechanisms of synergy. Lancet Oncol 2016; 16:e498-509. [PMID: 26433823 DOI: 10.1016/s1470-2045(15)00007-8] [Citation(s) in RCA: 639] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 05/20/2015] [Accepted: 05/21/2015] [Indexed: 12/11/2022]
Abstract
Checkpoint blockade immunotherapy has received mainstream attention as a result of striking and durable clinical responses in some patients with metastatic disease and a reasonable response rate in many tumour types. The activity of checkpoint blockade immunotherapy is not restricted to melanoma or lung cancer, and additional indications are expected in the future, with responses already reported in renal cancer, bladder cancer, and Hodgkin's lymphoma among many others. Additionally, the interactions between radiation and the immune system have been investigated, with several studies describing the synergistic effects on local and distant tumour control when radiation therapy is combined with immunotherapy. Clinical enthusiasm for this approach is strengthened by the many ongoing trials combining immunotherapy with definitive and palliative radiation. Herein, we discuss the biological and mechanistic rationale behind combining radiation with checkpoint blockade immunotherapy, with a focus on the preclinical data supporting this potentially synergistic combination. We explore potential hypotheses and important considerations for clinical trial designs. Finally, we reintroduce the notion of radiosensitising immunotherapy, akin to radiosensitising chemotherapy, as a potential definitive therapeutic modality.
Collapse
Affiliation(s)
- Andrew B Sharabi
- University of California, San Diego, Department of Radiation Medicine and Applied Sciences, La Jolla, CA, USA; Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Michael Lim
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA; Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Theodore L DeWeese
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA; Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA; The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Charles G Drake
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA; The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|