Discover the SciOpen Platform and Achieve Your Research Goals with Ease.
Search articles, authors, keywords, DOl and etc.
To compare neural damage induced by ultra-high dose rate FLASH radiotherapy (FLASH-RT) with that induced by conventional dose rate radiotherapy (CONV-RT) in healthy mice.
Eighty adult male C57BL/6J mice were divided into five groups: Sham, CONV-RT10Gy, CONV-RT20Gy, FLASH-RT10Gy, and FLASH-RT20Gy. Three days post-irradiation, morphological changes in neurons within the dentate gyrus (DG), CA1, and CA3 were observed using hematoxylin and eosin and Nissl staining. The malondialdehyde (MDA), reduced glutathione (GSH), glutathione peroxidase (GSH-PX), superoxide dismutase (SOD), catalase (CAT), and hydroxyl radical (OH−) levels were measured using assay kits. Quantitative reverse transcription PCR was used to assess interleukin (IL)-1β, IL-6, inducible nitric oxide synthase (iNOS), and tumor necrosis factor (TNF)-α mRNA expression levels in hippocampus. Immunofluorescence was employed to observe microglial activation in the DG.
Compared with Sham, CONV-RT10Gy and CONV-RT20Gy exhibited disorganized neuronal arrangements and blurred nucleoli in the DG; the number of Nissl body was reduced, but FLASH-RT10Gy and FLASH-RT20Gy alleviated these abnormalities. Moreover, FLASH-RT20Gy mitigated the upregulation of MDA and downregulation of GSH, GSH-PX, SOD, CAT, and OH− levels in the hippocampus of mice subjected to CONV-RT20Gy. Additionally, FLASH-RT20Gy attenuated the upregulation of IL-1β, IL-6, iNOS, and TNF-α mRNA levels in hippocampus of mice subjected to CONV-RT20Gy and diminished microglial activation in the DG.
FLASH-RT mitigate the structural and functional disruptions in hippocampal neurons induced by CONV-RT and alleviate oxidative stress and inflammation in hippocampal tissue by reducing microglial activation.
Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA A Cancer J Clin. 2024;74(1):12–49. https://doi.org/10.3322/caac.21820.
Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J Clin. 2021;71(3):209–249. https://doi.org/10.3322/caac.21660.
Lu ZH, Chen Y, Liu D, et al. The landscape of cancer research and cancer care in China. Nat Med. 2023;29(12):3022–3032. https://doi.org/10.1038/s41591-023-02655-3.
Zhou ZH, Guan BJ, Xia H, et al. Particle radiotherapy in the era of radioimmunotherapy. Cancer Lett. 2023;567:216268. https://doi.org/10.1016/j.canlet.2023.216268.
Yan SS, Ngoma TA, Ngwa W, et al. Global democratisation of proton radiotherapy. Lancet Oncol. 2023;24(6):e245–e254. https://doi.org/10.1016/S1470-2045(23)00184-5.
Manem VS, Taghizadeh-Hesary F. Advances in personalized radiotherapy. BMC Cancer. 2024;24(1):556. https://doi.org/10.1186/s12885-024-12317-3.
Lan JX, Ren YF, Liu YY, et al. A bibliometric analysis of radiation-induced brain injury: a research of the literature from 1998 to 2023. Discov Oncol. 2024;15(1):364. https://doi.org/10.1007/s12672-024-01223-6.
Li HN, Gong QY, Luo K. Biomarker-driven molecular imaging probes in radiotherapy. Theranostics. 2024;14(10):4127–4146. https://doi.org/10.7150/thno.97768.
Thariat J, Little MP, Zablotska LB, et al. Radiotherapy for non-cancer diseases: benefits and long-term risks. Int J Radiat Biol. 2024;100(4):505–526. https://doi.org/10.1080/09553002.2023.2295966.
Boire A, Brastianos PK, Garzia L, et al. Brain metastasis. Nat Rev Cancer. 2020;20(1):4–11. https://doi.org/10.1038/s41568-019-0220-y.
Wilke C, Grosshans D, Duman J, et al. Radiation-induced cognitive toxicity: pathophysiology and interventions to reduce toxicity in adults. Neuro Oncol. 2018;20(5):597–607. https://doi.org/10.1093/neuonc/nox195.
Dokic I, Meister S, Bojcevski J, et al. Neuroprotective effects of ultra-high dose rate FLASH Bragg peak proton irradiation. Int J Radiat Oncol Biol Phys. 2022;113(3):614–623. https://doi.org/10.1016/j.ijrobp.2022.02.020.
Favaudon V, Caplier L, Monceau V, et al. Ultrahigh dose-rate FLASH irradiation increases the differential response between normal and tumor tissue in mice. Sci Transl Med. 2014;6(245):245ra93. https://doi.org/10.1126/scitranslmed.3008973.
Allen BD, Acharya MM, Montay-Gruel P, et al. Maintenance of tight junction integrity in the absence of vascular dilation in the brain of mice exposed to ultra-high-dose-rate FLASH irradiation. Radiat Res. 2020;194(6):625–635. https://doi.org/10.1667/RADE-20-00060.1.
Shi ZS, Yu P, Lin WJ, et al. Microglia drive transient insult-induced brain injury by chemotactic recruitment of CD8+ T lymphocytes. Neuron. 2023;111(5):696–710.e9. https://doi.org/10.1016/j.neuron.2022.12.009.
Zhang Z, Jiang JR, He Y, et al. Pregabalin mitigates microglial activation and neuronal injury by inhibiting HMGB1 signaling pathway in radiation-induced brain injury. J Neuroinflammation. 2022;19(1):231. https://doi.org/10.1186/s12974-022-02596-7.
Turnquist C, Harris BT, Harris CC. Radiation-induced brain injury: current concepts and therapeutic strategies targeting neuroinflammation. Neurooncol Adv. 2020;2(1):vdaa057. https://doi.org/10.1093/noajnl/vdaa057.
Liu Q, Huang Y, Duan MY, et al. Microglia as therapeutic target for radiation-induced brain injury. Int J Mol Sci. 2022;23(15):8286. https://doi.org/10.3390/ijms23158286.
Cheng JP, Jiang JR, He BX, et al. A phase 2 study of thalidomide for the treatment of radiation-induced blood-brain barrier injury. Sci Transl Med. 2023;15(684):eabm6543. https://doi.org/10.1126/scitranslmed.abm6543.
Zhao XH, Cheng JP, Gui SS, et al. Amifostine-loaded nanocarrier traverses the blood-brain barrier and prevents radiation-induced brain injury. ACS Appl Mater Interfaces. 2023;15(12):15203–15219. https://doi.org/10.1021/acsami.3c00502.
Lin B, Huang D, Gao F, et al. Mechanisms of FLASH effect. Front Oncol. 2022;12:995612. https://doi.org/10.3389/fonc.2022.995612.
Lv YH, Lv Y, Wang Z, et al. FLASH radiotherapy: a promising new method for radiotherapy. Oncol Lett. 2022;24(6):419. https://doi.org/10.3892/ol.2022.13539.
Allen BD, Alaghband Y, Kramár EA, et al. Elucidating the neurological mechanism of the FLASH effect in juvenile mice exposed to hypofractionated radiotherapy. Neuro Oncol. 2023;25(5):927–939. https://doi.org/10.1093/neuonc/noac248.
Gao F, Yang YW, Zhu HY, et al. First demonstration of the FLASH effect with ultrahigh dose rate high-energy X-rays. Radiother Oncol. 2022;166:44–50. https://doi.org/10.1016/j.radonc.2021.11.004.
Yang YW, Wang JX, Gao F, et al. FLASH radiotherapy using high-energy X-rays: current status of PARTER platform in FLASH research. Radiother Oncol. 2023:109967. https://doi.org/10.1016/j.radonc.2023.109967.
Di Venanzio C, Marinelli M, Milani E, et al. Characterization of a synthetic single crystal diamond Schottky diode for radiotherapy electron beam dosimetry. Med Phys. 2013;40(2):021712. https://doi.org/10.1118/1.4774360.
Ciancaglioni I, Marinelli M, Milani E, et al. Dosimetric characterization of a synthetic single crystal diamond detector in clinical radiation therapy small photon beams. Med Phys. 2012;39(7):4493–4501. https://doi.org/10.1118/1.4729739.
Mascia AE, Daugherty EC, Zhang YB, et al. Proton FLASH radiotherapy for the treatment of symptomatic bone metastases: the FAST-01 nonrandomized trial. JAMA Oncol. 2023;9(1):62–69. https://doi.org/10.1001/jamaoncol.2022.5843.
Bourhis J, Sozzi WJ, Jorge PG, et al. Treatment of a first patient with FLASH-radiotherapy. Radiother Oncol. 2019;139:18–22. https://doi.org/10.1016/j.radonc.2019.06.019.
Sunnerberg JP, Zhang RX, Gladstone DJ, et al. Mean dose rate in ultra-high dose rate electron irradiation is a significant predictor for O2 consumption and H2O2 yield. Phys Med Biol. 2023;68(16):165014. https://doi.org/10.1088/1361-6560/ace877.
Schneider T, Fernandez-Palomo C, Bertho A, et al. Combining FLASH and spatially fractionated radiation therapy: the best of both worlds. Radiother Oncol. 2022;175:169–177. https://doi.org/10.1016/j.radonc.2022.08.004.
Zhu HY, Xie DH, Wang Y, et al. Comparison of intratumor and local immune response between MV X-ray FLASH and conventional radiotherapies. Clin Transl Radiat Oncol. 2022;38:138–146. https://doi.org/10.1016/j.ctro.2022.11.005.
Eggold JT, Chow S, Melemenidis S, et al. Abdominopelvic FLASH irradiation improves PD-1 immune checkpoint inhibition in preclinical models of ovarian cancer. Mol Cancer Therapeut. 2022;21(2):371–381. https://doi.org/10.1158/1535-7163.MCT-21-0358.
Vorhees CV, Vatner RE, Williams MT. Review of conventional and high dose rate brain radiation (FLASH): neurobehavioural, neurocognitive and assessment issues in rodent models. Clin Oncol. 2021;33(11):e482–e491. https://doi.org/10.1016/j.clon.2021.09.002.
Polevoy GG, Kumar DS, Daripelli S, et al. Flash therapy for cancer: a potentially new radiotherapy methodology. Cureus. 2023;15(10):e46928. https://doi.org/10.7759/cureus.46928.
This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).