Abstract
FLASH radiotherapy has the potential to improve both patient quality of life and outcomes by delivering radiation at ultrahigh dose rates to effectively target tumours while sparing healthy tissues. However, the differential sensitivity of healthy tissues versus tumours to FLASH radiotherapy remains unexplained. In this Perspective, we hypothesize that FLASH radiotherapy distinguishes healthy tissues from tumours based on subtle functional and structural biological differences. We identify commonalities present in the various healthy tissues that are spared by FLASH radiotherapy that might be lost during tumorigenesis. We also propose that a specific class of proteins, termed long-lived proteins, define a critical radiolytic target that are present in nearly every healthy tissue that is FLASH radiotherapy resistant yet are absent in tumours. We extend this structural hypothesis further by suggesting that tumour and extracellular matrix rigidity affects sensitivity to changes in radiotherapy dose rate, where more rigid and dense desmoplastic tumours are more sensitive to FLASH radiotherapy than those possessing more elasticity. Substantiating these concepts experimentally may provide a new and generalized mechanism of action of radiation effects and may therefore inform clinical trial designs by identifying those tumour subclasses expected to exhibit optimal responses to FLASH radiotherapy.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 /Â 30Â days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout



References
Jaffray, D. A. et al. Global task force on radiotherapy for cancer control. Lancet Oncol. 16, 1144â1146 (2015).
Mahvi, D. A., Liu, R., Grinstaff, M. W., Colson, Y. L. & Raut, C. P. Local cancer recurrence: the realities, challenges, and opportunities for new therapies. CA Cancer J. Clin. 68, 488â505 (2018).
Williams, C. P. et al. Importance of quality-of-life priorities and preferences surrounding treatment decision making in patients with cancer and oncology clinicians. Cancer 126, 3534â3541 (2020).
Favaudon, V. et al. Ultrahigh dose-rate FLASH irradiation increases the differential response between normal and tumor tissue in mice. Sci. Transl. Med. 6, 245ra293 (2014).
Inada, T., Nishio, H., Amino, S., Abe, K. & Saito, K. High dose-rate dependence of early skin reaction in mouse. Int. J. Radiat. Biol. Relat. Stud. Phys. Chem. Med. 38, 139â145 (1980).
Hendry, J. H., Moore, J. V., Hodgson, B. W. & Keene, J. P. The constant low oxygen concentration in all the target cells for mouse tail radionecrosis. Radiat. Res. 92, 172â181 (1982).
Limoli, C. L. & Vozenin, M.-C. Reinventing radiobiology in the light of FLASH radiotherapy. Annu. Rev. Cancer Biol. https://doi.org/10.1146/annurev-cancerbio-061421-022217 (2023).
Vozenin, M. C. et al. FLASH: New intersection of physics, chemistry, biology, and cancer medicine. Rev. Mod. Phys. 96, 1â50 (2024).
Loo, B. W. Jr et al. Navigating the critical translational questions for implementing FLASH in the clinic. Semin. Radiat. Oncol. 34, 351â364 (2024).
Bourhis, J. et al. Treatment of a first patient with FLASH-radiotherapy. Radiother. Oncol. 139, 18â22 (2019).
Gaide, O. et al. Comparison of ultra-high versus conventional dose rate radiotherapy in a patient with cutaneous lymphoma. Radiother. Oncol. https://doi.org/10.1016/j.radonc.2021.12.045 (2022).
Mascia, A. E. et al. Proton FLASH radiotherapy for the treatment of symptomatic bone metastases: the FAST-01 nonrandomized trial. JAMA Oncol. 9, 62â69 (2023).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT05524064 (2025).
Daugherty, E. C. et al. FLASH radiotherapy for the treatment of symptomatic bone metastases in the thorax (FAST-02): protocol for a prospective study of a novel radiotherapy approach. Radiat. Oncol. 19, 34 (2024).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT04986696 (2023).
US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/study/NCT05724875 (2025).
Kinj, R. et al. Randomized phase II selection trial of FLASH and conventional radiotherapy for patients with localized cutaneous squamous cell carcinoma or basal cell carcinoma: a study protocol. Clin. Transl. Radiat. Oncol. 45, 100743 (2024).
Allignet, B. et al. Long-term outcomes after definitive radiotherapy with modern techniques for unresectable soft tissue sarcoma. Radiother. Oncol. 173, 55â61 (2022).
Timmerman, R. D., Herman, J. & Cho, L. C. Emergence of stereotactic body radiation therapy and its impact on current and future clinical practice. J. Clin. Oncol. 32, 2847â2854 (2014).
Andratschke, N. et al. European Society for Radiotherapy and Oncology and European Organisation for Research and Treatment of Cancer consensus on re-irradiation: definition, reporting, and clinical decision making. Lancet Oncol. 23, e469âe478 (2022).
Verginadis, I. I. et al. FLASH proton reirradiation, with or without hypofractionation, reduces chronic toxicity in the normal murine intestine, skin, and bone. Radiother. Oncol. 205, 110744 (2025).
Clarke, M. et al. Effects of radiotherapy and of differences in the extent of surgery for early breast cancer on local recurrence and 15-year survival: an overview of the randomised trials. Lancet 366, 2087â2106 (2005).
Barrios, C. H., Reinert, T. & Werutsky, G. Global breast cancer research: moving forward. Am. Soc. Clin. Oncol. Educ. Book 38, 441â450 (2018).
Kacem, H. et al. Comparing radiolytic production of H2O2 and development of zebrafish embryos after ultra high dose rate exposure with electron and transmission proton beams. Radiother. Oncol. 175, 197â202 (2022).
Blain, G. et al. Proton irradiations at ultra-high dose rate vs. conventional dose rate: strong impact on hydrogen peroxide yield. Radiat. Res. 198, 318â324 (2022).
Kacem, H. et al. Modification of the microstructure of the CERN- CLEAR-VHEE beam at the picosecond scale modifies ZFE morphogenesis but has no impact on hydrogen peroxide production. Radiother. Oncol. 209, 110942 (2025).
Anderson, A. R. & Hart, E. J. Radiation chemistry of water with pulsed high intensity electron beams. J. Phys. Chem. 66, 70â75 (1962).
Sehested, K., Rasmussen, O. L. & Fricke, H. Rate constants of OH with HO2,O2â, and H2O2+ from hydrogen peroxide formation in pulse-irradiated oxygenated water. J. Phys. Chem. 72, 626â631 (1968).
Milligan, J. R., Aguilera, J. A. & Ward, J. F. Variation of single-strand break yield with scavenger concentration for plasmid DNA irradiated in aqueous solution. Radiat. Res. 133, 151 (1993).
Milligan, J. R., Arnold, A. D. & Ward, J. F. The effect of superhelical density on the yield of single-strand breaks in γ-irradiated plasmid DNA. Radiat. Res. 132, 69 (1992).
Kunz, L. V. et al. Plasmid DNA strand breaks are dose rate independent at clinically relevant proton doses and under biological conditions. Radiat. Res. 203, 214â222 (2024).
Perstin, A., Poirier, Y., Sawant, A. & Tambasco, M. Quantifying the DNA-damaging effects of FLASH irradiation with plasmid DNA. Int. J. Radiat. Oncol. Biol. Phys. https://doi.org/10.1016/j.ijrobp.2022.01.049 (2022).
Wanstall, H. C. et al. VHEE FLASH sparing effect measured at CLEAR, CERN with DNA damage of pBR322 plasmid as a biological endpoint. Sci. Rep. 14, 14803 (2024).
Froidevaux, P. et al. FLASH irradiation does not induce lipid peroxidation in lipids micelles and liposomes. Radiat. Phys. Chem. https://doi.org/10.1016/j.radphyschem.2022.110733 (2023).
Grilj, V. et al. Average dose rate is the primary determinant of lipid peroxidation in liposome membranes exposed to pulsed electron FLASH beam. Radiat. Phys. Chem. https://doi.org/10.1016/j.radphyschem.2024.111887 (2024).
Portier, L. et al. Differential remodeling of the oxylipin pool after FLASH versus conventional dose-rate irradiation in vitro and in vivo. Int. J. Radiat. Oncol. Biol. Phys. 119, 1481â1492 (2024).
Gupta, S. et al. A novel platform for evaluating dose rate effects on oxidative damage to peptides: toward a high-throughput method to characterize the mechanisms underlying the FLASH effect. Radiat. Res. 200, 523â530 (2023).
Martinez-Rovira, I. et al. Infrared microspectroscopy to elucidate the underlying biomolecular mechanisms of FLASH radiotherapy. Radiother. Oncol. 196, 110238 (2024).
Jansen, J. et al. Ex vivo brain MRI to assess conventional and FLASH brain irradiation effects. Radiother. Oncol. 208, 110894 (2025).
Montay-Gruel, P. et al. Long-term neurocognitive benefits of FLASH radiotherapy driven by reduced reactive oxygen species. Proc. Natl Acad. Sci. USA 116, 10943â10951 (2019).
Allen, B. D. et al. Elucidating the neurological mechanism of the FLASH effect in juvenile mice exposed to hypofractionated radiotherapy. Neuro. Oncol. https://doi.org/10.1093/neuonc/noac248 (2022).
Hall, E. J. & Giaccia, A. J. Radiobiology for the Radiologist 7th edn (Wolters Kluwer Health/Lippincott Williams & Wilkins, 2012).
Joiner, M. C. & van der Kogel, A. J. Basic Clinical Radiobiology 6th edn (CRC, 2025).
Balentova, S. & Adamkov, M. Molecular, cellular and functional effects of radiation-induced brain injury: a review. Int. J. Mol. Sci. 16, 27796â27815 (2015).
Yarnold, J. & Brotons, M. C. Pathogenetic mechanisms in radiation fibrosis. Radiother. Oncol. 97, 149â161 (2010).
Verginadis, I. I. et al. Radiotherapy toxicities: mechanisms, management, and future directions. Lancet 405, 338â352 (2025).
Levy, K. et al. Abdominal FLASH irradiation reduces radiation-induced gastrointestinal toxicity for the treatment of ovarian cancer in mice. Sci. Rep. 10, 21600 (2020).
Diffenderfer, E. S. et al. Design, implementation, and in vivo validation of a novel proton FLASH radiation therapy system. Int. J. Radiat. Oncol. Biol. Phys. 106, 440â448 (2020).
Ruan, J. L. et al. Irradiation at ultra-high (FLASH) dose rates reduces acute normal tissue toxicity in the mouse gastrointestinal system. Int. J. Radiat. Oncol. Biol. Phys. 111, 1250â1261 (2021).
Kim, M. M. et al. Comparison of FLASH proton entrance and the spread-out bragg peak dose regions in the sparing of mouse intestinal crypts and in a pancreatic tumor model. Cancers https://doi.org/10.3390/cancers13164244 (2021).
Singers Sorensen, B. et al. In vivo validation and tissue sparing factor for acute damage of pencil beam scanning proton FLASH. Radiother. Oncol. 167, 109â115 (2022).
Montay-Gruel, P. et al. Irradiation in a flash: unique sparing of memory in mice after whole brain irradiation with dose rates above 100Gy/s. Radiother. Oncol. 124, 365â369 (2017).
Bohlen, T. T. et al. Normal tissue sparing by FLASH as a function of single fraction dose: a quantitative analysis. Int. J. Radiat. Oncol. Biol. Phys. https://doi.org/10.1016/j.ijrobp.2022.05.038 (2022).
Horst, F. et al. Dose and dose rate dependence of the tissue sparing effect at ultra-high dose rate studied for proton and electron beams using the zebrafish embryo model. Radiother. Oncol. 194, 110197 (2024).
Kimmel, C. B., Ballard, W. W., Kimmel, S. R., Ullmann, B. & Schilling, T. F. Stages of embryonic development of the zebrafish. Dev. Dyn. 203, 253â310 (1995).
Fouillade, C. et al. FLASH irradiation spares lung progenitor cells and limits the incidence of radio-induced senescence. Clin. Cancer Res. 26, 1497â1506 (2020).
Velalopoulou, A. et al. FLASH proton radiotherapy spares normal epithelial and mesenchymal tissues while preserving sarcoma response. Cancer Res. 81, 4808â4821 (2021).
Kim, K. et al. FLASH proton radiation therapy mitigates inflammatory and fibrotic pathways and preserves cardiac function in a preclinical mouse model of radiation-induced heart disease. Int. J. Radiat. Oncol. Biol. Phys. 119, 1234â1247 (2024).
Alaghband, Y. et al. Neuroprotection of radiosensitive juvenile mice by ultra-high dose rate FLASH irradiation. Cancers https://doi.org/10.3390/cancers12061671 (2020).
Montay-Gruel, P. et al. X-rays can trigger the FLASH effect: ultra-high dose-rate synchrotron light source prevents normal brain injury after whole brain irradiation in mice. Radiother. Oncol. 129, 582â588 (2018).
Montay-Gruel, P. et al. Hypofractionated FLASH-RT as an effective treatment against glioblastoma that reduces neurocognitive side effects in mice. Clin. Cancer Res. 27, 775â784 (2021).
Alexander, T. C. et al. Behavioral effects of focal irradiation in a juvenile murine model. Radiat. Res. 189, 605â617 (2018).
Chambers, S. M. et al. Aging hematopoietic stem cells decline in function and exhibit epigenetic dysregulation. PLoS Biol. 5, e201 (2007).
Booth, L. N. & Brunet, A. The aging epigenome. Mol. Cell 62, 728â744 (2016).
Lopez-Otin, C., Blasco, M. A., Partridge, L., Serrano, M. & Kroemer, G. Hallmarks of aging: an expanding universe. Cell 186, 243â278 (2023).
Montay-Gruel, P. et al. Ultra-high-dose-rate FLASH irradiation limits reactive gliosis in the brain. Radiat. Res. 194, 636â645 (2020).
Simmons, D. A. et al. Reduced cognitive deficits after FLASH irradiation of whole mouse brain are associated with less hippocampal dendritic spine loss and neuroinflammation. Radiother. Oncol. 139, 4â10 (2019).
Kacem, H., Almeida, A., Cherbuin, N. & Vozenin, M. C. Understanding the FLASH effect to unravel the potential of ultra-high dose rate irradiation. Int. J. Radiat. Biol. 98, 506â516 (2022).
Allen, B. D. 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. 194, 625â635 (2020).
Barghouth, P. G. et al. FLASH-RT does not affect chromosome translocations and junction structures beyond that of CONV-RT dose-rates. Radiother. Oncol. 188, 109906 (2023).
Withers, H. R. in Advances in Radiation Biology Vol. 5 (eds Lett, J. T. & Adler, H.) 241â271 (Academic, 1975).
Royce, T. J. et al. Tumor control probability modeling and systematic review of the literature of stereotactic body radiation therapy for prostate cancer. Int. J. Radiat. Oncol. Biol. Phys. 110, 227â236 (2021).
Chabi, S. et al. Ultra-high-dose-rate FLASH and conventional-dose-rate irradiation differentially affect human acute lymphoblastic leukemia and normal hematopoiesis. Int. J. Radiat. Oncol. Biol. Phys. 109, 819â829 (2021).
Leavitt, R. J. et al. Acute hypoxia does not alter tumor sensitivity to FLASH radiation therapy. Int. J. Radiat. Oncol. Biol. Phys. https://doi.org/10.1016/j.ijrobp.2024.02.015 (2024).
Gray, L. H., Conger, A. D., Ebert, M., Hornsey, S. & Scott, O. C. The concentration of oxygen dissolved in tissues at the time of irradiation as a factor in radiotherapy. Br. J. Radiol. 26, 638â648 (1953).
Wilson, J. D., Hammond, E. M., Higgins, G. S. & Petersson, K. Ultra-high dose rate (FLASH) radiotherapy: silver bullet or foolâs gold? Front. Oncol. 9, 1563 (2019).
Jansen, J. et al. Does FLASH deplete oxygen? Experimental evaluation for photons, protons, and carbon ions. Med. Phys. 48, 3982â3990 (2021).
El Khatib, M. et al. Ultrafast tracking of oxygen dynamics during proton FLASH. Int. J. Radiat. Oncol. Biol. Phys. 113, 624â634 (2022).
Cao, X. et al. Quantification of oxygen depletion during FLASH irradiation in vitro and in vivo. Int. J. Radiat. Oncol. Biol. Phys. 111, 240â248 (2021).
Grilj, V. et al. In vivo measurements of change in tissue oxygen level during irradiation reveal novel dose rate dependence. Radiother. Oncol. 201, 110539 (2024).
Jin, J.-Y. et al. Ultra-high dose rate effect on circulating immune cells: a potential mechanism for FLASH effect? Radiother. Oncol. 149, 55â62 (2020).
Iturri, L. et al. Proton FLASH radiation therapy and immune infiltration: evaluation in an orthotopic glioma rat model. Int. J. Radiat. Oncol. Biol. Phys. 116, 655â665 (2023).
Almeida, A. et al. Dosimetric and biologic intercomparison between electron and proton FLASH beams. Radiother. Oncol. 190, 109953 (2024).
Almeida, A. et al. Antitumor effect by either FLASH or conventional dose rate irradiation involves equivalent immune responses. Int. J. Radiat. Oncol. Biol. Phys. 118, 1110â1122 (2024).
Padilla, O. et al. Immune response following FLASH and conventional radiation in diffuse midline glioma. Int. J. Radiat. Oncol. Biol. Phys. 119, 1248â1260 (2024).
Cunningham, S. et al. FLASH proton pencil beam scanning irradiation minimizes radiation-induced leg contracture and skin toxicity in mice. Cancers https://doi.org/10.3390/cancers13051012 (2021).
Ni, H. et al. FLASH radiation reprograms lipid metabolism and macrophage immunity and sensitizes medulloblastoma to CAR-T cell therapy. Nat. Cancer https://doi.org/10.1038/s43018-025-00905-6 (2025).
Geraghty, A. C. et al. Immunotherapy-related cognitive impairment after CAR T cell therapy in mice. Cell 188, 3238â3258.e3225 (2025).
Kim, Y.-E. et al. Effects of ultra-high doserate FLASH irradiation on the tumor microenvironment in Lewis lung carcinoma: role of myosin light chain. Int. J. Radiat. Oncol. Biol. Phys. 109, 1440â1453 (2021).
Yang, L. et al. Pathophysiological responses in rat and mouse models of radiation-induced brain injury. Mol. Neurobiol. 54, 1022â1032 (2017).
Toyama, B. H. & Hetzer, M. W. Protein homeostasis: live long, wonât prosper. Nat. Rev. Mol. Cell Biol. 14, 55â61 (2013).
Toyama, B. H. et al. Identification of long-lived proteins reveals exceptional stability of essential cellular structures. Cell 154, 971â982 (2013).
Krishna, S. et al. Identification of long-lived proteins in the mitochondria reveals increased stability of the electron transport chain. Dev. Cell 56, 2952â2965.e2959 (2021).
Savas, J. N., Toyama, B. H., Xu, T., Yates, J. R. 3rd & Hetzer, M. W. Extremely long-lived nuclear pore proteins in the rat brain. Science 335, 942 (2012).
Toyama, B. H. et al. Visualization of long-lived proteins reveals age mosaicism within nuclei of postmitotic cells. J. Cell Biol. 218, 433â444 (2019).
Arrojo, E. D. R. et al. Age mosaicism across multiple scales in adult tissues. Cell Metab. 30, 343â351.e343 (2019).
Bomba-Warczak, E. & Savas, J. N. Long-lived mitochondrial proteins and why they exist. Trends Cell Biol. 32, 646â654 (2022).
Limoli, C. L., Laposa, R. & Cleaver, J. E. DNA replication arrest in XP variant cells after UV exposure is diverted into an Mre11-dependent recombination pathway by the kinase inhibitor wortmannin. Mutat. Res. 510, 121â129 (2002).
Straub, M. et al. Distinct nitrogen isotopic compositions of healthy and cancerous tissue in mice brain and head and neck micro-biopsies. BMC Cancer 21, 805 (2021).
Carlos, A. R. & Allamand, V. Editorial: extracellular matrix in homeostasis and cancer. Front. Genet. 13, 1107969 (2022).
Cambria, E. et al. Linking cell mechanical memory and cancer metastasis. Nat. Rev. Cancer 24, 216â228 (2024).
Toulemonde, M., Surdutovich, E. & Solovâyov, A. V. Temperature and pressure spikes in ion-beam cancer therapy. Phys. Rev. E 80, 031913 (2009).
Bjegovic, K. et al. 4D in vivo dosimetry for a FLASH electron beam using radiation-induced acoustic imaging. Phys. Med. Biol. https://doi.org/10.1088/1361-6560/ad4950 (2024).
Lascaud, J. & Parodi, K. On the potential biological impact of radiation-induced acoustic emissions during ultra-high dose rate electron radiotherapy: a preliminary study. Phys. Med. Biol. https://doi.org/10.1088/1361-6560/acb9ce (2023).
Rohrer Bley, C. et al. Dose- and volume-limiting late toxicity of FLASH radiotherapy in cats with squamous cell carcinoma of the nasal planum and in mini pigs. Clin. Cancer Res. 28, 3814â3823 (2022).
Lascaud, J., Radler, M., Rohrer Bley, C., Vozenin, M. C. & Parodi, K. Retrospective study on the resonance of thermoacoustic emissions and their possible biological implications in cats treated with electron FLASH beams. Phys. Med. Biol. https://doi.org/10.1088/1361-6560/adb679 (2025).
Abolfath, R., Baikalov, A., Bartzsch, S., Afshordi, N. & Mohan, R. The effect of non-ionizing excitations on the diffusion of ion species and inter-track correlations in FLASH ultra-high dose rate radiotherapy. Phys. Med. Biol. https://doi.org/10.1088/1361-6560/ac69a6 (2022).
Novak, U. & Kaye, A. H. Extracellular matrix and the brain: components and function. J. Clin. Neurosci. 7, 280â290 (2000).
Dunsmore, S. E. & Rannels, D. E. Extracellular matrix biology in the lung. Am. J. Physiol. 270, L3âL27 (1996).
Hussey, G. S., Keane, T. J. & Badylak, S. F. The extracellular matrix of the gastrointestinal tract: a regenerative medicine platform. Nat. Rev. Gastroenterol. Hepatol. 14, 540â552 (2017).
Feng, X. et al. Targeting extracellular matrix stiffness for cancer therapy. Front. Immunol. 15, 1467602 (2024).
Huang, J. et al. Extracellular matrix and its therapeutic potential for cancer treatment. Signal Transduct. Target. Ther. 6, 153 (2021).
Tinganelli, W. et al. FLASH with carbon ions: tumor control, normal tissue sparing, and distal metastasis in a mouse osteosarcoma model. Radiother. Oncol. 175, 185â190 (2022).
Liljedahl, E. et al. Long-term anti-tumor effects following both conventional radiotherapy and FLASH in fully immunocompetent animals with glioblastoma. Sci. Rep. 12, 12285 (2022).
Attygalle, A. D. et al. The 5th edition of the World health organization classification of mature lymphoid and stromal tumors â an overview and update. Leuk. Lymphoma 65, 413â429 (2024).
Gao, F. et al. First demonstration of the FLASH effect with ultrahigh dose rate high-energy X-rays. Radiother. Oncol. 166, 44â50 (2022).
Barty, C. P. J. et al. Design, construction, and test of compact, distributed-charge, X-band accelerator systems that enable image-guided, VHEE FLASH radiotherapy. Front. Phys. 12, 1472759 (2024).
Behjati, S. et al. Mutational signatures of ionizing radiation in second malignancies. Nat. Commun. 7, 12605 (2016).
Vozenin, M. C., Bourhis, J. & Durante, M. Towards clinical translation of FLASH radiotherapy. Nat. Rev. Clin. Oncol. 19, 791â803 (2022).
Sorensen, B. S. et al. Pencil beam scanning proton FLASH maintains tumor control while normal tissue damage is reduced in a mouse model. Radiother. Oncol. 175, 178â184 (2022).
Sorensen, B. S. et al. Proton FLASH: impact of dose rate and split dose on acute skin toxicity in a murine model. Int. J. Radiat. Oncol. Biol. Phys. 120, 265â275 (2024).
Mascia, A. et al. Impact of multiple beams on the FLASH effect in soft tissue and skin in mice. Int. J. Radiat. Oncol. Biol. Phys. 118, 253â261 (2024).
Bohlen, T. T. et al. The minimal FLASH sparing effect needed to compensate the increase of radiobiological damage due to hypofractionation for late-reacting tissues. Med. Phys. 49, 7672â7682 (2022).
Limoli, C. L. et al. The sparing effect of FLASH-RT on synaptic plasticity is maintained in mice with standard fractionation. Radiother. Oncol. 186, 109767 (2023).
Chowdhury, P. et al. Proton FLASH radiotherapy ameliorates radiation-induced salivary gland dysfunction and oral mucositis and increases survival in a mouse model of head and neck cancer. Mol. Cancer Ther. 23, 877â889 (2024).
Alaghband, Y. et al. Uncovering the protective neurologic mechanisms of hypofractionated FLASH radiotherapy. Cancer Res. Commun. 3, 725â737 (2023).
Acknowledgements
We would like to thank V. Favaudon (Inserm), M. Hetzer (ISTA), J. Lascaud (LMU), R. Abolfath (Howard U) and A. Durham (HUG) for fruitful scientific discussions as well as the junior fellows in our teams A. Almeida (Unige) and O. Drayson (UCI) for their support in preparing figures.
Author information
Authors and Affiliations
Contributions
The authors contributed equally to all aspects of the article.
Corresponding author
Ethics declarations
Competing interests
M.-C.V. declares one research grant from Varian, Siemens Healthineers dedicated to FLASH preclinical research, one research grant from IBA dedicated to FLASH preclinical research and one research grant from Roche dedicated to radio-immunotherapy. P.M.-G. and P.T. declare no competing interests. C.L.L. declares receiving consulting fees from IBA dedicated to FLASH developments.
Peer review
Peer review information
Nature Reviews Cancer thanks Loredana Marcu and the other, anonymous, reviewers for their contribution to the peer review of this work.
Additional information
Publisherâs note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary information
Glossary
- Acoustic cavitation
-
The formation, growth and implosive collapse of bubbles in a liquid due to pressure fluctuations induced by sound waves, typically ultrasound.
- Acoustic oscillation
-
The periodic variation in pressure, particle velocity and other acoustic quantities in a medium as a sound wave propagates through it.
- Desmoplastic tumour
-
A tumour that develops fibrous connective tissue and adhesions that increase stiffness.
- DNA damage response
-
A series of cellular signalling events following DNA damage to promote repair.
- Dosimetric tools
-
Devices and methods used to measure and quantify the amount of radiation delivered to a specific area, ensuring accuracy and safety in radiotherapy and other radiation applications.
- Extracellular matrix
-
A cellular stromal components composed of macromolecules (that is, proteins, carbohydrates and lipids) and minerals, which provide structural and biochemical support to cells.
- Fibrosis
-
Progressive and chronic alteration of a tissue due to pathological wound healing characterized by the replacement of a normal parenchymal tissue by connective tissue.
- Free radical recombination
-
A chemical reaction in which two free radicals combine to form a stable, non-radical molecule.
- Ionizing radiation
-
Particles or electromagnetic waves with sufficient energy to induce the ionization of atoms it interacts with, and in the context of external beam radiotherapy, these are often X-rays, protons, electrons or carbon ions.
- Isodose
-
An equivalent dose given by two different irradiation modalities.
- Proteostasis
-
The biological processes involved in the synthesis, folding, trafficking and degradation of proteins required for the proper maintenance of cellular functions.
- Radiation chemistry
-
The study of the chemical effects and reactions induced by the absorption of ionizing radiation in matter.
- Radiobiology
-
A branch of science studying the interactions of ionizing radiation with biological tissues and organisms.
- Radiolytic oxygen depletion
-
The consumption of oxygen by radiation-induced chemical reactions in aqueous systems, producing ROS and reducing overall oxygen level.
- Radioresistance
-
Intrinsic or adaptive capacity of cells, tissues, organs or organisms to overcome the detrimental effects of ionizing radiation.
- Therapeutic index
-
The measurement of treatment efficacy relative to its side effects.
- Tumour clamping
-
An experimental procedure used to restrict blood flow to a tumour, inducing hypoxic or ischaemic conditions.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Vozenin, MC., Montay-Gruel, P., Tsoutsou, P. et al. Mechanisms, challenges and opportunities for FLASH radiotherapy in cancer. Nat Rev Cancer (2025). https://doi.org/10.1038/s41568-025-00878-9
Accepted:
Published:
DOI: https://doi.org/10.1038/s41568-025-00878-9