1
|
Kuipers ME, van Liefferinge F, van der Wal E, Rovituso M, Slats AM, Hiemstra PS, Van Doorn-Wink KC. Effect of FLASH proton therapy on primary bronchial epithelial cell organoids. Clin Transl Radiat Oncol 2025; 52:100927. [PMID: 39968050 PMCID: PMC11833640 DOI: 10.1016/j.ctro.2025.100927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 01/21/2025] [Accepted: 01/28/2025] [Indexed: 02/20/2025] Open
Abstract
Purpose The effects of conventional (CONV) and FLASH proton therapy on primary bronchial epithelial cell (PBEC) organoids from individuals with chronic obstructive pulmonary disease (COPD) were investigated. The primary objective was to compare the effect of FLASH and CONV on COPD PBEC organoids with a focus on DNA damage, organoid formation, and gene expression. Methods PBECs were obtained from six COPD donors, cultured as three-dimensional (3D) organoids and exposed to 2 and 8 Gy CONV and FLASH proton radiation at the Holland Proton Therapy Center. DNA damage was assessed by γH2AX staining. Organoid formation capacity was assessed by counting the organoids formed after reseeding irradiated cells at 24 h and 7 days. Bulk RNA sequencing (RNAseq) and qPCR analyses were performed to identify pathways and differences in the radiation response. Results γH2AX foci analysis showed a significant dose-dependent increase in DNA damage at 1 h for both CONV and FLASH treatments, without differences between the two modalities. Organoid formation assays revealed a dose-dependent decrease in organoid formation capacity at 24 h for both treatments. At 7 days, 2 Gy FLASH-treated samples showed significantly reduced organoid formation compared to 2 Gy CONV (p = 0.008). RNAseq identified CONV and FLASH-induced changes in expression of DNA-damage response and apoptosis pathway genes. A dose-dependent upregulation of MDM2, GDF15, DDB2, BAX, P21, AEN and a decrease in MKi67 expression was confirmed by qPCR analysis. Conclusion No significant differences were found in DNA damage or gene expression profiles between CONV and FLASH. The organoid formation assay showed a prolonged detrimental effect in the FLASH-treated organoids, suggesting a more complex interaction of FLASH with lung epithelial cells. The results of this study contribute to the advancement of robust in vitro human lung models for investigating the mechanisms of action of FLASH, potentially facilitating the treatment of NSCLC patients with proton FLASH therapy.
Collapse
Affiliation(s)
- Merian E. Kuipers
- Leiden University Medical Center (LUMC), Department of Pulmonology, C02-Q, Albinusdreef 2 2333 ZA Leiden, the Netherlands
| | - Floriane van Liefferinge
- Leiden University Medical Center (LUMC), Department of Pulmonology, C02-Q, Albinusdreef 2 2333 ZA Leiden, the Netherlands
| | - Ernst van der Wal
- Holland Proton Therapy Center (HollandPTC), Huismansingel 4 2629 JH Delft, the Netherlands
| | - Marta Rovituso
- Holland Proton Therapy Center (HollandPTC), Huismansingel 4 2629 JH Delft, the Netherlands
| | - Annelies M. Slats
- Leiden University Medical Center (LUMC), Department of Pulmonology, C02-Q, Albinusdreef 2 2333 ZA Leiden, the Netherlands
| | - Pieter S. Hiemstra
- Leiden University Medical Center (LUMC), Department of Pulmonology, C02-Q, Albinusdreef 2 2333 ZA Leiden, the Netherlands
| | - Krista C.J. Van Doorn-Wink
- Holland Proton Therapy Center (HollandPTC), Huismansingel 4 2629 JH Delft, the Netherlands
- Leiden University Medical Center (LUMC), Department of Radiotherapy, K01-P, Albinusdreef 2 2333 ZA Leiden, the Netherlands
| |
Collapse
|
2
|
Li XK, Amirkhanyan Z, Grebinyk A, Gross M, Komar Y, Riemer F, Asoyan A, Boonpornprasert P, Borchert P, Davtyan H, Dmytriiev D, Frohme M, Hoffmann A, Krasilnikov M, Loisch G, Lotfi Z, Müller F, Schmitz M, Obier F, Oppelt A, Philipp S, Richard C, Vashchenko G, Villani D, Worm S, Stephan F. Demonstration of ultra-high dose rate electron irradiation at FLASH lab@PITZ. Phys Med Biol 2025; 70:055010. [PMID: 39907068 DOI: 10.1088/1361-6560/adb276] [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: 10/16/2024] [Accepted: 01/30/2025] [Indexed: 02/06/2025]
Abstract
Objective.The photo injector test facility at DESY in Zeuthen (PITZ) is building up an R&D platform, known as FLASHlab@PITZ, for systematically studying the FLASH effect in cancer treatment with its high-brightness electron beams, which can provide a uniquely large dose parameter range for radiation experiments. In this paper, we demonstrate the capabilities by experiments with a reduced parameter range on a startup beamline and study the potential performance of the full beamline by simulations.Approach.To measure the dose, Gafchromic films are installed both in front of and after the samples; Monte Carlo simulations are conducted to predict the dose distribution during beam preparation and help understand the dose distribution inside the sample. Plasmid DNA is irradiated under various doses at conventional and ultra-high dose rate (UHDR) to study the DNA damage by radiations. Start-to-end simulations are performed to verify the performance of the full beamline.Main results.On the startup beamline, reproducible irradiation has been established with optimized electron beams and the delivered dose distributions have been measured with Gafchromic films and compared to FLUKA simulations. The functionality of this setup has been further demonstrated in biochemical experiments at conventional dose rate of 0.05 Gy s-1and UHDR of several 105 Gy s-1and a varying dose up to 60 Gy, with the UHDR experiments finished within a single RF pulse (less than 1 millisecond); the observed conformation yields of the irradiated plasmid DNA revealed its dose-dependent radiation damage. The upgrade to the full FLASHlab@PITZ beamline is justified by simulations with homogeneous radiation fields generated by both pencil beam scanning and scattering beams.Significance.With the demonstration of UHDR irradiation and the simulated performance of the new beamline, FLASHlab@PITZ will serve as a powerful platform for studying the FLASH effects in cancer treatment.
Collapse
Affiliation(s)
- X-K Li
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - Z Amirkhanyan
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - A Grebinyk
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
- Technical University of Applied Sciences Wildau, 15745 Wildau, Germany
| | - M Gross
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - Y Komar
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
- Technical University of Applied Sciences Wildau, 15745 Wildau, Germany
| | - F Riemer
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - A Asoyan
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - P Boonpornprasert
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - P Borchert
- Technical University of Applied Sciences Wildau, 15745 Wildau, Germany
| | - H Davtyan
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - D Dmytriiev
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - M Frohme
- Technical University of Applied Sciences Wildau, 15745 Wildau, Germany
| | - A Hoffmann
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - M Krasilnikov
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - G Loisch
- Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
| | - Z Lotfi
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - F Müller
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - M Schmitz
- Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
| | - F Obier
- Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
| | - A Oppelt
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - S Philipp
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - C Richard
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - G Vashchenko
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - D Villani
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - S Worm
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - F Stephan
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| |
Collapse
|
3
|
Kristensen L, Rohrer S, Hoffmann L, Præstegaard LH, Ankjærgaard C, Andersen CE, Kanouta E, Johansen JG, Sahlertz M, Nijkamp J, Poulsen PR, Sørensen BS. Electron vs proton FLASH radiation on murine skin toxicity. Radiother Oncol 2025:110796. [PMID: 39983873 DOI: 10.1016/j.radonc.2025.110796] [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: 11/15/2024] [Revised: 01/22/2025] [Accepted: 02/12/2025] [Indexed: 02/23/2025]
Abstract
BACKGROUND AND PURPOSE Dose-response modification of FLASH has previously been established for acute skin toxicity in protons. This study used a similar experimental setup to quantify the dose-response modification of electron FLASH irradiation for acute skin- and late fibrotic toxicity in mice. The setup similarity enabled quantitative comparison of the acute skin response for electrons to protons. METHOD Female unanaesthetised C3D2F1 mice were restrained with the right hindleg fixated and submerged in a water bath for horizontal electron irradiation at 16 MeV. Mice were randomised in groups of varying single doses (19.4-57.6 Gy) and irradiated with either 0.162 Gy/s conventional (CONV) or 233 Gy/s FLASH dose rate using 8-10 mice per group. Acute skin toxicity was assessed daily from the 8th to the 28th day post-irradiation. The same mice were kept for a fibrotic assay of leg extension assessment done biweekly until 52 weeks post-irradiation. The dose-modifying factor (DMF) of FLASH was quantified from dose-response curves. RESULTS AND DISCUSSION Electron FLASH irradiated mice showed a considerable skin-sparing effect with a DMF of 1.45-1.54 and a smaller fibrotic-sparing effect with a DMF of 1.15. The development of acute skin toxicity was similar between CONV and FLASH groups with biological equivalent doses based on the DMF. The acute response of the electron irradiations was similar to previous reports on protons. CONCLUSION Despite apparent differences, e.g. average and instantaneous dose rates, the acute skin toxicity of electron beams and previously published proton beams were remarkably similar regarding both biological response and quantified acute skin DMFs.
Collapse
Affiliation(s)
- Line Kristensen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Experimental Clinical Oncology, Aarhus University Hospital, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Oncology, Aarhus University Hospital, Aarhus, Denmark.
| | - Sky Rohrer
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Lone Hoffmann
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | | | | | - Claus E Andersen
- DTU Health Tech, Technical University of Denmark, Roskilde, Denmark
| | - Eleni Kanouta
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jacob Graversgaard Johansen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Morten Sahlertz
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jasper Nijkamp
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Per Rugaard Poulsen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Brita Singers Sørensen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Experimental Clinical Oncology, Aarhus University Hospital, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
4
|
Ni H, Reitman ZJ, Zou W, Akhtar MN, Paul R, Huang M, Zhang D, Zheng H, Zhang R, Ma R, Ngo G, Zhang L, Diffenderfer ES, Motlagh SAO, Kim MM, Minn AJ, Dorsey JF, Foster JB, Metz J, Koumenis C, Kirsch DG, Gong Y, Fan Y. FLASH radiation reprograms lipid metabolism and macrophage immunity and sensitizes medulloblastoma to CAR-T cell therapy. NATURE CANCER 2025:10.1038/s43018-025-00905-6. [PMID: 39910249 DOI: 10.1038/s43018-025-00905-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/07/2025] [Indexed: 02/07/2025]
Abstract
FLASH radiotherapy holds promise for treating solid tumors given the potential lower toxicity in normal tissues but its therapeutic effects on tumor immunity remain largely unknown. Using a genetically engineered mouse model of medulloblastoma, we show that FLASH radiation stimulates proinflammatory polarization in tumor macrophages. Single-cell transcriptome analysis shows that FLASH proton beam radiation skews macrophages toward proinflammatory phenotypes and increases T cell infiltration. Furthermore, FLASH radiation reduces peroxisome proliferator-activated receptor-γ (PPARγ) and arginase 1 expression and inhibits immunosuppressive macrophage polarization under stimulus-inducible conditions. Mechanistically, FLASH radiation abrogates lipid oxidase expression and oxidized low-density lipid generation to reduce PPARγ activity, while standard radiation induces reactive oxygen species-dependent PPARγ activation in macrophages. Notably, FLASH radiotherapy improves infiltration and activation of chimeric antigen receptor (CAR) T cells and sensitizes medulloblastoma to GD2 CAR-T cell therapy. Thus, FLASH radiotherapy reprograms macrophage lipid metabolism to reverse tumor immunosuppression. Combination FLASH-CAR radioimmunotherapy may offer exciting opportunities for solid tumor treatment.
Collapse
Affiliation(s)
- Haiwei Ni
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Zachary J Reitman
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - Wei Zou
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Md Naushad Akhtar
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ritama Paul
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Menggui Huang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Duo Zhang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Hao Zheng
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ruitao Zhang
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ruiying Ma
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Gina Ngo
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Lin Zhang
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
| | - Eric S Diffenderfer
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Michele M Kim
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Andy J Minn
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
- Mark Foundation Center for Immunotherapy, Immune Signaling, and Radiation, University of Pennsylvania, Philadelphia, PA, USA
| | - Jay F Dorsey
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jessica B Foster
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - James Metz
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
- Mark Foundation Center for Immunotherapy, Immune Signaling, and Radiation, University of Pennsylvania, Philadelphia, PA, USA
| | - David G Kirsch
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA.
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada.
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| | - Yanqing Gong
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA.
- Mark Foundation Center for Immunotherapy, Immune Signaling, and Radiation, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Bell BI, Velten C, Pennock M, Kang M, Tanaka KE, Selvaraj B, Bookbinder A, Koba W, Vercellino J, English J, Małachowska B, Pandey S, Duddempudi PK, Yang Y, Shajahan S, Hasan S, Choi JI, Simone CB, Yang WL, Tomé WA, Lin H, Guha C. Whole Abdominal Pencil Beam Scanned Proton FLASH Increases Acute Lethality. Int J Radiat Oncol Biol Phys 2025; 121:493-505. [PMID: 39299552 DOI: 10.1016/j.ijrobp.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/29/2024] [Accepted: 09/02/2024] [Indexed: 09/22/2024]
Abstract
PURPOSE Ultrahigh dose-rate FLASH radiation therapy has emerged as a modality that promises to reduce normal tissue toxicity while maintaining tumor control. Previous studies of gastrointestinal toxicity using passively scattered FLASH proton therapy (PRT) have, however, yielded mixed results, suggesting that the requirements for gastrointestinal sparing by FLASH are an open question. Furthermore, the more clinically relevant pencil beam scanned (PBS) FLASH PRT has not yet been assessed in this context, despite differences in the spatiotemporal dose-rate distributions compared with passively scattered PRT. Here, to our knowledge, we provide the first report on the effects of PBS FLASH PRT on acute gastrointestinal injury in mice after whole abdominal irradiation. METHODS AND MATERIALS Whole abdominal irradiation was performed on C57BL/6J mice using the entrance channel of the Bragg curve of a 250 MeV PBS proton beam at field-averaged dose rates of 0.6 Gy/s for conventional (CONV) and 80 to 100 Gy/s for FLASH PRT. A 2D strip ionization chamber array was used to measure the dose and dose rate for each mouse. Survival was assessed at 14 Gy. Intestines were harvested and processed as Swiss rolls for analysis using a novel artificial intelligence-based crypt assay to quantify crypt regeneration 4 days after irradiation. RESULTS Survival was significantly reduced after 14 Gy FLASH PRT compared with CONV (P < .001). Our artificial intelligence-based crypt assays demonstrated no significant difference in intestinal crypts/cm or crypt depth between groups 4 days after irradiation. Furthermore, we found no significant difference in 5-ethynyl-2'-deoxyuridine+ cells/crypt or Olfactomedin4+ intestinal stem cells with FLASH relative to CONV PRT. CONCLUSIONS Overall, our data demonstrate significantly impaired survival after abdominal PBS FLASH PRT without apparent differences in intestinal histology 4 days after irradiation.
Collapse
Affiliation(s)
- Brett I Bell
- Departments of Radiation Oncology, Albert Einstein College of Medicine, Bronx, New York; Departments of Radiation Oncology and Pathology, Albert Einstein College of Medicine, Bronx, New York
| | - Christian Velten
- Departments of Radiation Oncology, Albert Einstein College of Medicine, Bronx, New York
| | - Michael Pennock
- Departments of Radiation Oncology, Albert Einstein College of Medicine, Bronx, New York
| | | | - Kathryn E Tanaka
- Departments of Radiation Oncology and Pathology, Albert Einstein College of Medicine, Bronx, New York
| | | | | | - Wade Koba
- Department of Radiology, Albert Einstein College of Medicine, Bronx, New York
| | - Justin Vercellino
- Departments of Radiation Oncology, Albert Einstein College of Medicine, Bronx, New York; Departments of Radiation Oncology and Pathology, Albert Einstein College of Medicine, Bronx, New York
| | - Jeb English
- Departments of Radiation Oncology, Albert Einstein College of Medicine, Bronx, New York; Departments of Radiation Oncology and Pathology, Albert Einstein College of Medicine, Bronx, New York
| | - Beata Małachowska
- Departments of Radiation Oncology, Albert Einstein College of Medicine, Bronx, New York
| | - Sanjay Pandey
- Departments of Radiation Oncology, Albert Einstein College of Medicine, Bronx, New York
| | | | - Yunjie Yang
- New York Proton Center, New York, New York; Departments of Medical Physics and Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Shahin Shajahan
- Departments of Radiation Oncology, Albert Einstein College of Medicine, Bronx, New York
| | | | - J Isabelle Choi
- New York Proton Center, New York, New York; Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Charles B Simone
- New York Proton Center, New York, New York; Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Weng-Lang Yang
- Departments of Radiation Oncology, Albert Einstein College of Medicine, Bronx, New York
| | - Wolfgang A Tomé
- Departments of Radiation Oncology, Albert Einstein College of Medicine, Bronx, New York; Department of Neurology, Albert Einstein College of Medicine, Bronx, New York
| | - Haibo Lin
- Departments of Radiation Oncology, Albert Einstein College of Medicine, Bronx, New York; New York Proton Center, New York, New York; Departments of Medical Physics and Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Chandan Guha
- Departments of Radiation Oncology, Albert Einstein College of Medicine, Bronx, New York; Departments of Radiation Oncology and Pathology, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
6
|
Bietenbeck V, Bäcker CM, Wulff J, Timmermann B, Bäumer C. Proof-of-principle of 3D-printed track-end detectors for dosimetry in proton therapy. Med Phys 2025; 52:737-741. [PMID: 39556581 PMCID: PMC11788257 DOI: 10.1002/mp.17515] [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: 07/16/2024] [Revised: 10/01/2024] [Accepted: 10/10/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND Dosimetric equipment in particle therapy (PT) is associated with high costs. There is a lack of versatile, tissue-equivalent detectors suitable for in-vivo dosimetry. Faraday-cup (FC) type detectors are sensitive to stopped protons, that is, to track-ends (TEs). They experience a renaissance in PT as they can cope with high dose rates. Owing to their simple functional principle, production of FC could benefit from the dynamic technological developments in additive manufacturing of sensors. PURPOSE To build FC-type detectors for PT by standard 3D-printing. This study seeks to build an integrating, single-channel (SC) FC for replacement of a traditional FC and a2 × 2 $2\times 2$ array of FC elements indicating the feasibility of a spatially resolving detector. METHODS Samples of FCs were produced with a dual-extruder 3D-printer with polylactic-acid filaments, which contained graphite in the conductive parts of the detector. Production was optimized in terms of materials and printing temperature. Samples were characterized by electrical tests and non-destructive 3D x-ray imaging. Beam tests were conducted at a clinical PT machine. RESULTS Operational FC-type detectors for proton fields were printed. The detected charge of the SC FC corresponded qualitatively to the one of a traditional FC. A2 × 2 $2\times 2$ FC array was fabricated in a single run. There was a linear relationship between the response of the individual FC elements and the machine output. CONCLUSIONS 3D-printing is a viable method for producing low-cost, tissue-equivalent, FC-type detectors for PT. They could potentially be used as TE detectors in anthropomorphic phantoms.
Collapse
Affiliation(s)
- Vicky Bietenbeck
- West German Proton Therapy Centre EssenEssenGermany
- West German Cancer Center (WTZ)University Hospital EssenEssenGermany
| | - Claus Maximilian Bäcker
- West German Proton Therapy Centre EssenEssenGermany
- West German Cancer Center (WTZ)University Hospital EssenEssenGermany
| | - Jörg Wulff
- West German Proton Therapy Centre EssenEssenGermany
- West German Cancer Center (WTZ)University Hospital EssenEssenGermany
| | - Beate Timmermann
- West German Proton Therapy Centre EssenEssenGermany
- West German Cancer Center (WTZ)University Hospital EssenEssenGermany
- Department of Particle TherapyUniversity Hospital EssenEssenGermany
- German Cancer Consortium (DKTK)EssenGermany
| | - Christian Bäumer
- West German Proton Therapy Centre EssenEssenGermany
- West German Cancer Center (WTZ)University Hospital EssenEssenGermany
- German Cancer Consortium (DKTK)EssenGermany
- Department of PhysicsTU Dortmund UniversityDortmundGermany
| |
Collapse
|
7
|
Wang Y, Wang H, Hu J, Chai J, Luan J, Li J, Xu Q. FLASH radiotherapy: mechanisms, nanotherapeutic strategy and future development. NANOSCALE ADVANCES 2025; 7:711-721. [PMID: 39781242 PMCID: PMC11705069 DOI: 10.1039/d4na00753k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/21/2024] [Indexed: 01/12/2025]
Abstract
Ultra-high dose-rate (FLASH) radiotherapy serves as an ideal procedure to treat tumors efficiently without harming normal tissues and has demonstrated satisfactory antitumor effects in multiple animal tumor models. However, the biological mechanisms of FLASH radiotherapy have not yet been fully elucidated, and the small number of devices delivering FLASH dose rate has limited its wide application. This review summarizes the possible biological mechanisms and antitumor effects of FLASH radiotherapy, its application in nanotherapeutic strategy, as well as its challenges and future development. Furthermore, some valuable guidance for promoting the progress of FLASH radiotherapy in nanotherapeutic strategies are provided.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College Wuhu China
| | - Huifang Wang
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College Wuhu China
| | - Jiawei Hu
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College Wuhu China
| | - Jingjing Chai
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College Wuhu China
| | - Jiajie Luan
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College Wuhu China
| | - Jie Li
- Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China Mianyang China
| | - Qingwen Xu
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College Wuhu China
| |
Collapse
|
8
|
Verginadis II, Velalopoulou A, Kim MM, Kim K, Paraskevaidis I, Bell B, Oliaei Motlagh SA, Karaj A, Banerjee E, Finesso G, Assenmacher CA, Radaelli E, Lu J, Lin Y, Putt ME, Diffenderfer ES, Guha C, Qin L, Metz JM, Maity A, Cengel KA, Koumenis C, Busch TM. FLASH proton reirradiation, with or without hypofractionation, reduces chronic toxicity in the normal murine intestine, skin, and bone. Radiother Oncol 2025; 205:110744. [PMID: 39880309 DOI: 10.1016/j.radonc.2025.110744] [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: 07/05/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND AND PURPOSE The normal tissue sparing afforded by FLASH radiotherapy is being intensely investigated for potential clinical translation. Here, we studied the effects of FLASH proton radiotherapy (F-PRT) in the reirradiation setting, with or without hypofractionation. Chronic toxicities in three murine models of normal tissue toxicity including the intestine, skin, and bone were investigated. MATERIALS AND METHODS In studies of the intestine, single-dose irradiation was performed with 12 Gy of standard proton RT (S-PRT), followed by a second dose of 12 Gy of F-PRT or S-PRT. Additionally, a hypofractionation scheme was applied in the reirradiation setting (3 x 6.4 Gy of F-PRT or S-PRT, given every 48 hrs). In studies of skin/bone of the murine leg, 15 Gy of S-PRT was followed by hypofractionated reirradiation with F-PRT or S-PRT (3 x 11 Gy). RESULTS Compared to reirradiation with S-PRT, F-PRT induced less intestinal fibrosis and collagen deposition that was accompanied by significantly increased survival rate, demonstrating its protective effects on intestinal tissues in the reirradiation setting. In previously irradiated leg tissues, reirradiation with hypofractionated F-PRT created transient dermatitis that fully resolved in contrast to reirradiation with hypofractionated S-PRT. Lymphedema was also alleviated after a second course of radiation with F-PRT, along with significant reductions in the accumulation of fibrous connective tissue in the skin, compared to mice reirradiated with S-PRT. The delivery of a second course of fractionated S-PRT induced tibial fractures in 83.3% of the mice, whereas only 20% of mice reirradiated with F-PRT presented with fractures. CONCLUSION These studies provide the first evidence of the sparing effects of F-PRT in the setting of hypofractionated reirradiation. The results support FLASH as highly relevant to the reirradiation regimen where it exhibits significant potential to minimize chronic complications for patients undergoing RT.
Collapse
Affiliation(s)
- Ioannis I Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anastasia Velalopoulou
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michele M Kim
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kyle Kim
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ioannis Paraskevaidis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Brett Bell
- Department of Radiation Oncology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA
| | - Seyyedeh Azar Oliaei Motlagh
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Antoneta Karaj
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Esha Banerjee
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Giovanni Finesso
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Charles-Antoine Assenmacher
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Enrico Radaelli
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jiawei Lu
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuewei Lin
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mary E Putt
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eric S Diffenderfer
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chandan Guha
- Department of Radiation Oncology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ling Qin
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James M Metz
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amit Maity
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Keith A Cengel
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Theresa M Busch
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Verginadis II, Citrin DE, Ky B, Feigenberg SJ, Georgakilas AG, Hill-Kayser CE, Koumenis C, Maity A, Bradley JD, Lin A. Radiotherapy toxicities: mechanisms, management, and future directions. Lancet 2025; 405:338-352. [PMID: 39827884 DOI: 10.1016/s0140-6736(24)02319-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 10/09/2024] [Accepted: 10/18/2024] [Indexed: 01/22/2025]
Abstract
For over a century, radiotherapy has revolutionised cancer treatment. Technological advancements aim to deliver high doses to tumours with increased precision while minimising off-target effects to organs at risk. Despite advancements such as image-guided, high-precision radiotherapy delivery, long-term toxic effects on healthy tissues remain a great clinical challenge. In this Review, we summarise common mechanisms driving acute and long-term side-effects and discuss monitoring strategies for radiotherapy survivors. We explore ways to mitigate toxic effects through novel technologies and proper patient selection and counselling. Additionally, we address policies and management strategies to minimise the severity and impact of toxicity during and after treatment. Finally, we examine the potential advantages of emerging technologies and innovative approaches to improve conformity, accuracy, and minimise off-target effects.
Collapse
Affiliation(s)
- Ioannis I Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Deborah E Citrin
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Bonnie Ky
- Department of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Steven J Feigenberg
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexandros G Georgakilas
- Department of Physics, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Christine E Hill-Kayser
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Cancer Center, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amit Maity
- Department of Radiation Oncology, University of Utah Health, Salt Lake City, UT, USA
| | - Jeffrey D Bradley
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander Lin
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
10
|
Liu K, Titt U, Esplen N, Connell L, Konradsson E, Yang M, Wang X, Takaoka T, Li Z, Koong AC, Mitra D, Mohan R, Loo BW, Lin SH, Schüler E. Discordance in Acute Gastrointestinal Toxicity between Synchrotron-Based Proton and Linac-based Electron Ultra-High Dose Rate Irradiation. Int J Radiat Oncol Biol Phys 2025:S0360-3016(25)00058-6. [PMID: 39862897 DOI: 10.1016/j.ijrobp.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/16/2024] [Accepted: 01/12/2025] [Indexed: 01/27/2025]
Abstract
PURPOSE Proton FLASH has been investigated using cyclotron and synchrocyclotron beamlines but not synchrotron beamlines. We evaluated the impact of dose rate [ultra-high vs conventional (CONV)] and beam configuration [shoot-through (S-T) vs spread-out Bragg peak (SOBP)] on acute radiation-induced gastrointestinal toxicity (RIGIT) in mice. We also compared RIGIT between synchrotron-based protons and linac-based electrons with matched mean dose rates. METHODS AND MATERIALS We administered abdominal irradiation (12-14 Gy single fraction) to female C57BL/6J mice with an 87-MeV synchrotron-based proton beamline (2-cm-diameter field size as a lateral beam). Dose rates were 0.2 Gy/s (S-T pCONV), 0.3 Gy/s (SOBP pCONV), 150 Gy/s (S-T pFLASH), and 230 Gy/s (SOBP pFLASH). RIGIT was assessed by the jejunal regenerating crypt assay and survival. We also compared responses to proton (pFLASH and pCONV) with responses to electron CONV (eCONV, 0.4 Gy/s) and electron-beam FLASH (188-205 Gy/s). RESULTS The number of regenerating jejunal crypts at each matched dose was lowest for pFLASH (similar between S-T and SOBP), greater and similar between pCONV (S-T and SOBP) and eCONV, and greatest for electron-beam FLASH. Correspondingly, mice that received pFLASH SOBP had the lowest survival rates (50% at 50 days), followed by pFLASH S-T (80%), and pCONV SOBP (90%), but 100% of mice receiving pCONV S-T survived (log-rank P = .047 for the 4 groups). CONCLUSIONS Our findings are consistent with an increase in RIGIT after synchrotron-based pFLASH versus pCONV. This negative proton-specific FLASH effect versus linac-based electron irradiation underscores the importance of understanding the physical and biological factors that will allow safe and effective clinical translation.
Collapse
Affiliation(s)
- Kevin Liu
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, Texas; The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Uwe Titt
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, Texas; The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Nolan Esplen
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Luke Connell
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, Texas; The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Elise Konradsson
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ming Yang
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, Texas; The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Xiaochun Wang
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Takeshi Takaoka
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, Texas; Particle Therapy Division, Hitachi America Ltd, Houston, Texas
| | - Ziyi Li
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Albert C Koong
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Devarati Mitra
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Radhe Mohan
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, Texas; The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Billy W Loo
- Department of Radiation Oncology and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Steven H Lin
- The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas; Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Emil Schüler
- Department of Radiation Physics, University of Texas MD Anderson Cancer Center, Houston, Texas; The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas.
| |
Collapse
|
11
|
Xiao Y, Benedict S, Cui Y, Glide-Hurst C, Graves S, Jia X, Kry SF, Li H, Lin L, Matuszak M, Newpower M, Paganetti H, Qi XS, Roncali E, Rong Y, Sgouros G, Simone CB, Sunderland JJ, Taylor PA, Tchelebi L, Weldon M, Zou JW, Wuthrick EJ, Machtay M, Le QT, Buchsbaum JC. Embracing the Future of Clinical Trials in Radiation Therapy: An NRG Oncology CIRO Technology Retreat Whitepaper on Pioneering Technologies and AI-Driven Solutions. Int J Radiat Oncol Biol Phys 2025:S0360-3016(25)00057-4. [PMID: 39848295 DOI: 10.1016/j.ijrobp.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/20/2024] [Accepted: 01/12/2025] [Indexed: 01/25/2025]
Abstract
This white paper examines the potential of pioneering technologies and artificial intelligence-driven solutions in advancing clinical trials involving radiation therapy. As the field of radiation therapy evolves, the integration of cutting-edge approaches such as radiopharmaceutical dosimetry, FLASH radiation therapy, image guided radiation therapy, and artificial intelligence promises to improve treatment planning, patient care, and outcomes. Additionally, recent advancements in quantum science, linear energy transfer/relative biological effect, and the combination of radiation therapy and immunotherapy create new avenues for innovation in clinical trials. The paper aims to provide an overview of these emerging technologies and discuss their challenges and opportunities in shaping the future of radiation oncology clinical trials. By synthesizing knowledge from experts across various disciplines, this white paper aims to present a foundation for the successful integration of these innovations into radiation therapy research and practice, ultimately enhancing patient outcomes and revolutionizing cancer care.
Collapse
Affiliation(s)
- Ying Xiao
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Stanley Benedict
- Department of Radiation Oncology, University of California at Davis, Comprehensive Cancer Center, Davis, California
| | - Yunfeng Cui
- Department of Radiation Oncology, Duke University, Durham, North Carolina
| | - Carri Glide-Hurst
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin
| | - Stephen Graves
- Department of Radiology, Division of Nuclear Medicine, University of Iowa, Iowa City, Iowa
| | - Xun Jia
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland
| | - Stephen F Kry
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Heng Li
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland
| | - Liyong Lin
- Department of Radiation Oncology, Emory University, Atlanta, Georgia
| | - Martha Matuszak
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Mark Newpower
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Harald Paganetti
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - X Sharon Qi
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, California
| | - Emilie Roncali
- Department of Radiology, University of California at Davis, Davis, California
| | - Yi Rong
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - George Sgouros
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland
| | | | - John J Sunderland
- Department of Radiology, Division of Nuclear Medicine, University of Iowa, Iowa City, Iowa
| | - Paige A Taylor
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Leila Tchelebi
- Department of Radiation Oncology, Northwell Health, Mt. Kisco, New York
| | - Michael Weldon
- Department of Radiation Oncology, The Ohio State University Medical Center, Columbus, Ohio
| | - Jennifer W Zou
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Evan J Wuthrick
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Mitchell Machtay
- Department of Radiation Oncology, Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania
| | - Quynh-Thu Le
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Jeffrey C Buchsbaum
- Division of Cancer Treatment and Diagnosis, Radiation Research Program, National Cancer Institute, Bethesda, Maryland.
| |
Collapse
|
12
|
Katsuki S, Minami K, Oniwa K, Yagi M, Shimizu S, Hamatani N, Takashina M, Kanai T, Ogawa K. Ultra-high dose rate (FLASH) carbon ion irradiation inhibited immune suppressive protein expression on Pan02 cell line. JOURNAL OF RADIATION RESEARCH 2025; 66:97-102. [PMID: 39724928 PMCID: PMC11753840 DOI: 10.1093/jrr/rrae091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/22/2024] [Indexed: 12/28/2024]
Abstract
Recently, ultra-high dose rate (> 40 Gy/s, uHDR; FLASH) radiation therapy (RT) has attracted interest, because the FLASH effect that is, while a cell-killing effect on cancer cells remains, the damage to normal tissue could be spared has been reported. This study aimed to compare the immune-related protein expression on cancer cells after γ-ray, conventionally used dose rate (Conv) carbon ion (C-ion), and uHDR C-ion. B16F10 murine melanoma and Pan02 murine pancreas cancer were irradiated with γ-ray at Osaka University and with C-ion at Osaka HIMAK. The dose rates at 1.16 Gy/s for Conv and 380 Gy/s for uHDR irradiation. The expressed calreticulin (CRT), major histocompatibility complex class (MHC)-I, and programmed cell death 1 ligand (PD-L1) were evaluated by flow cytometry. Western blotting and PCR were utilized to evaluate endoplasmic reticulum (ER) stress, DNA damage, and its repair pathway. CRT, MHC-I on B16F10 was also increased by irradiation, while only C-ion increased MHC-I on Pan02. Notably, PD-L1 on B16F10 was increased after irradiation with both γ-ray and C-ion, while uHDR C-ion suppressed the expression of PD-L1 on Pan02. The present study indicated that uHDR C-ion has a different impact on the repair pathway of DNA damage and ER than the Conv C-ion. This is the first study to show the immune-related protein expressions on cancer cells after uHDR C-ion irradiation.
Collapse
Affiliation(s)
- Shohei Katsuki
- Department of Medical Physics and Engineering, Osaka University Graduate School of Medicine, 1-7 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kazumasa Minami
- Department of Medical Physics and Engineering, Osaka University Graduate School of Medicine, 1-7 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Karin Oniwa
- Department of Medical Physics and Engineering, Osaka University Graduate School of Medicine, 1-7 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masashi Yagi
- Department of Carbon Ion Radiotherapy, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shinichi Shimizu
- Department of Carbon Ion Radiotherapy, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Noriaki Hamatani
- Department of Medical Physics, Osaka Heavy Ion Therapy Center, 3-1-10 Otemae, Chuo-ku, Osaka-city, Osaka, 540-0008, Japan
| | - Masaaki Takashina
- Department of Medical Physics, Osaka Heavy Ion Therapy Center, 3-1-10 Otemae, Chuo-ku, Osaka-city, Osaka, 540-0008, Japan
| | - Tatsuaki Kanai
- Department of Medical Physics, Osaka Heavy Ion Therapy Center, 3-1-10 Otemae, Chuo-ku, Osaka-city, Osaka, 540-0008, Japan
| | - Kazuhiko Ogawa
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
13
|
Manring HR, Fleming JL, Meng W, Gamez M, Blakaj DM, Chakravarti A. FLASH Radiotherapy: From In Vivo Data to Clinical Translation. Hematol Oncol Clin North Am 2025:S0889-8588(24)00153-9. [PMID: 39828472 DOI: 10.1016/j.hoc.2024.11.008] [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: 01/22/2025]
Abstract
Delivery of radiotherapy (RT) at ultra-high dose rates or FLASH radiotherapy (FLASH-RT) is an emerging treatment option for patients with cancer that could increase survival outcomes and quality of life. In vivo data across a multitude of normal tissues and associated tumors have been published demonstrating the FLASH effect while bringing attention to the need for additional research. Combination of FLASH-RT with other treatment options including spatially fractionated RT, immunotherapy, and usage in the setting of reirradiation could also provide additional benefit. Phase I clinical trials have shown promising results, yet research is warranted before routine clinical use of FLASH-RT.
Collapse
Affiliation(s)
- Heather R Manring
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Jessica L Fleming
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Wei Meng
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Mauricio Gamez
- Department of Radiation Oncology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Dukagjin M Blakaj
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Arnab Chakravarti
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|
14
|
Wang Y, Qi SN, Bi N, Li YX. FLASH radiotherapy combined with immunotherapy: From biological mechanisms to blockbuster therapeutics. Transl Oncol 2025; 51:102183. [PMID: 39613524 PMCID: PMC11629542 DOI: 10.1016/j.tranon.2024.102183] [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: 06/23/2024] [Revised: 09/19/2024] [Accepted: 10/29/2024] [Indexed: 12/01/2024] Open
Abstract
FLASH ultra-high dose rate radiotherapy (RT) can effectively exert the protective effect on normal tissue and reduce the risk of treatment-related toxicity, without compromising the killing effect on tumor tissue, resulting in a significant differential biological effect between tumor control and normal tissue damage, namely the FLASH effect. To date, the precise biological details of the FLASH effect remain uncertain. The currently mainstream mechanisms proposed by the academic community include the transient oxygen depletion hypothesis, free radical hypothesis, immune protection hypothesis, and DNA integrity hypothesis, which have attracted increasing attention in recent years. Based on these theoretical principles and numerous investigations on the FLASH effect in vivo and in vitro, the combined application of FLASH and immune checkpoint inhibitors (ICIs) has been considered synergistic and potentially practical. The primary underlying basis is that FLASH might actively preserve the number and function of circulating immune cells, thereby enhancing the efficacy of immune cell-mediated immunotherapy. Meanwhile, FLASH RT could activate the tumor immune microenvironment and transform "cold'' tumors into ''hot'' ones, consequently boosting local and systemic anti-tumor immunity and expanding the therapeutic benefits of ICIs. Moreover, FLASH might attenuate immunoinflammatory responses and minimize the incidence of radiation-related adverse events, allowing for the potentially safer and promising clinical application of combing FLASH RT with ICI therapy. Nevertheless, data on this treatment modality is currently lacking, and several barriers remain to be addressed, including the logistical bottlenecks, technical hurdles, limited availability, and unclear biological mechanisms. Further research is warranted in the future.
Collapse
Affiliation(s)
- Yu Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing 100021, China
| | - Shu-Nan Qi
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing 100021, China.
| | - Nan Bi
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing 100021, China; State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Ye-Xiong Li
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing 100021, China.
| |
Collapse
|
15
|
Rothwell B, Bertolet A, Schuemann J. Proton FLASH-arc therapy (PFAT): A feasibility study for meeting FLASH dose-rate requirements in the clinic. Radiother Oncol 2025; 202:110623. [PMID: 39528113 DOI: 10.1016/j.radonc.2024.110623] [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: 04/30/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND AND PURPOSE Proton arc therapy and FLASH radiotherapy (FLASH-RT) each offer unique advantages in proton therapy. However, clinical translation of FLASH-RT faces challenges in defining and delivering high dose rates. We propose the use of proton FLASH-arc therapy (PFAT) to leverage the benefits of arc while addressing FLASH delivery concerns by spatially fractionating dose delivery to healthy tissue. MATERIALS AND METHODS Treatment plans for an abdominal phantom and a clinical brain case were designed in OpenTPS, using monoenergetic beams within a 360-degree gantry rotation. Beams were optimized to achieve target coverage while maximizing spatial fractionation in non-target regions. The temporal dose delivery to healthy-tissue voxels, or in specified organs-at-risk (OARs), was constrained via selective spot removal in the beamlets matrix. The dose, LET, number of spots per voxel, and voxel-wise average dose rate were calculated for each PFAT plan and compared to a corresponding IMPT scenario. RESULTS PFAT plans demonstrated comparable dose conformity to IMPT, with LET hotspots shifted towards the target center. The number of spots influencing healthy-tissue voxels was reduced, leading to regions of substantially higher dose rates in many points outside the target. OAR dose-rate optimization in the brain plan resulted in dose rates exceeding 40 Gy/s in the majority of points in the brainstem. CONCLUSION The PFAT technique combines the advantages of FLASH and arc therapy, providing improved LET distributions and enhanced biological effect in the target, while achieving high dose rates in healthy tissue, thus reducing healthy tissue damage. This feasibility study demonstrates the capability of PFAT, setting the foundation for further optimization and application in diverse patient cases and complex geometries.
Collapse
Affiliation(s)
- Bethany Rothwell
- Physics Division, Department of Radiation Oncology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, USA.
| | - Alejandro Bertolet
- Physics Division, Department of Radiation Oncology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, USA.
| | - Jan Schuemann
- Physics Division, Department of Radiation Oncology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Hart A, Dudzic JP, Clarke JW, Eby J, Perlman SJ, Bazalova-Carter M. High-throughput, low-cost FLASH: irradiation of Drosophila melanogaster with low-energy X-rays using time structures spanning conventional and ultrahigh dose rates. JOURNAL OF RADIATION RESEARCH 2024; 65:836-844. [PMID: 39422537 PMCID: PMC11629999 DOI: 10.1093/jrr/rrae079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/16/2024] [Indexed: 10/19/2024]
Abstract
FLASH radiotherapy is an emerging technique in radiation oncology that may improve clinical outcomes by reducing normal tissue toxicities. The physical radiation characteristics needed to induce the radiobiological benefits of FLASH are still an active area of investigation. To determine the dose rate, range of doses and delivery time structure necessary to trigger the FLASH effect, Drosophila melanogaster were exposed to ultrahigh dose rate (UHDR) or conventional radiotherapy dose rate (CONV) 120-kVp X-rays. A conventional X-ray tube outfitted with a shutter system was used to deliver 17- to 44-Gy doses to third-instar D. melanogaster larvae at both UHDR (210 Gy/s) and CONV (0.2-0.4 Gy/s) dose rates. The larvae were then tracked through development to adulthood and scored for eclosion and lifespan. Larvae exposed to UHDR eclosed at higher rates and had longer median survival as adults compared to those treated with CONV at the same doses. Eclosion rates at 24 Gy were 68% higher for the UHDR group (P < 0.05). Median survival from 22 Gy was >22 days for UHDR and 17 days for CONV (P < 0.01). Two normal tissue-sparing effects were observed for D. melanogaster irradiated with UHDR 120-kVp X-rays. The effects appeared only at intermediate doses and may be useful in establishing the dose range over which the benefits of FLASH can be obtained. This work also demonstrates the usefulness of a high-throughput fruit fly model and a low-cost X-ray tube system for radiobiological FLASH research.
Collapse
Affiliation(s)
- Alexander Hart
- Department of Physics and Astronomy, University of Victoria, 3800 Finnerty Road, Victoria, BC V8P 5C2, Canada
| | - Jan P Dudzic
- Department of Biology, University of Victoria, 3800 Finnerty Road, Victoria, BC V8P 5C2, Canada
| | - Jameson W Clarke
- Department of Biology, University of Victoria, 3800 Finnerty Road, Victoria, BC V8P 5C2, Canada
| | - Jonathan Eby
- Institute of Biomedical Engineering, University of Toronto, 164 College St. Toronto, Ontario M5S 3E2, Canada
| | - Steve J Perlman
- Department of Biology, University of Victoria, 3800 Finnerty Road, Victoria, BC V8P 5C2, Canada
| | - Magdalena Bazalova-Carter
- Department of Physics and Astronomy, University of Victoria, 3800 Finnerty Road, Victoria, BC V8P 5C2, Canada
| |
Collapse
|
17
|
Drayson OGG, Melemenidis S, Katila N, Viswanathan V, Kramár EA, Zhang R, Kim R, Ru N, Petit B, Dutt S, Manjappa R, Ramish Ashraf M, Lau B, Soto L, Skinner L, Yu AS, Surucu M, Maxim PG, Zebadua-Ballasteros P, Wood MA, Montay-Gruel P, Baulch JE, Vozenin MC, Loo BW, Limoli CL. A multi-institutional study to investigate the sparing effect after whole brain electron FLASH in mice: Reproducibility and temporal evolution of functional, electrophysiological, and neurogenic endpoints. Radiother Oncol 2024; 201:110534. [PMID: 39293721 PMCID: PMC11588524 DOI: 10.1016/j.radonc.2024.110534] [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: 05/27/2024] [Revised: 08/13/2024] [Accepted: 09/03/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND AND PURPOSE Ultra-high dose-rate radiotherapy (FLASH) has been shown to mitigate normal tissue toxicities associated with conventional dose rate radiotherapy (CONV) without compromising tumor killing in preclinical models. A prominent challenge in preclinical radiation research, including FLASH, is validating both the physical dosimetry and the biological effects across multiple institutions. MATERIALS AND METHODS We previously demonstrated dosimetric reproducibility of two different electron FLASH devices at separate institutions using standardized phantoms and dosimeters. In this study, tumor-free adult female mice were given 10 Gy whole brain FLASH and CONV irradiation at both institutions and evaluated for the reproducibility and temporal evolution of multiple neurobiological endpoints. RESULTS FLASH sparing of behavioral performance on novel object recognition (4 months post-irradiation) and of electrophysiologic long-term potentiation (LTP, 5 months post-irradiation) was reproduced between institutions. Differences between FLASH and CONV on the endpoints of hippocampal neurogenesis (Sox2, doublecortin), neuroinflammation (microglial activation), and electrophysiology (LTP) were not observed at early times (48 h to 2 weeks), but recovery of immature neurons by 3 weeks was greater with FLASH. CONCLUSION In summary, we demonstrated reproducible FLASH sparing effects on the brain between two different beams at two different institutions with validated dosimetry. FLASH sparing effects on the endpoints evaluated manifested at later but not the earliest time points.
Collapse
Affiliation(s)
- Olivia G G Drayson
- Department of Radiation Oncology, University of California, Irvine, CA 92697, USA
| | - Stavros Melemenidis
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nikita Katila
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Vignesh Viswanathan
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Enikö A Kramár
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Richard Zhang
- Department of Radiation Oncology, University of California, Irvine, CA 92697, USA
| | - Rachel Kim
- Department of Radiation Oncology, University of California, Irvine, CA 92697, USA
| | - Ning Ru
- Department of Radiation Oncology, University of California, Irvine, CA 92697, USA
| | - Benoit Petit
- Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Suparna Dutt
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rakesh Manjappa
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - M Ramish Ashraf
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Brianna Lau
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Luis Soto
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lawrie Skinner
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Amu S Yu
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Murat Surucu
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Peter G Maxim
- Department of Radiation Oncology, University of California, Irvine, CA 92697, USA
| | - Paola Zebadua-Ballasteros
- Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Switzerland; Laboratorio de Fisica Medica, Instituto Nacional de Neurología y Neurocirugía MVS, México City 14269, Mexico
| | - Marcelo A Wood
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Pierre Montay-Gruel
- Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Switzerland; Radiation Oncology Department, Iridium Netwerk, Wilrijk, Antwerp, Belgium; Antwerp Research in Radiation Oncology (AReRO), Centre for Oncological Research (CORE), University of Antwerp, Antwerp, Belgium
| | - Janet E Baulch
- Department of Radiation Oncology, University of California, Irvine, CA 92697, USA
| | - Marie-Catherine Vozenin
- Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Switzerland.
| | - Billy W Loo
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
18
|
Liu K, Holmes S, Khan AU, Hooten B, DeWerd L, Schüler E, Beddar S. Development of novel ionization chambers for reference dosimetry in electron flash radiotherapy. Med Phys 2024; 51:9275-9289. [PMID: 39311014 DOI: 10.1002/mp.17425] [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: 02/20/2024] [Revised: 08/10/2024] [Accepted: 09/10/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Reference dosimetry in ultra-high dose rate (UHDR) beamlines is significantly hindered by limitations in conventional ionization chamber design. In particular, conventional chambers suffer from severe charge collection efficiency (CCE) degradation in high dose per pulse (DPP) beams. PURPOSE The aim of this study was to optimize the design and performance of parallel plate ion chambers for use in UHDR dosimetry applications, and evaluate their potential as reference class chambers for calibration purposes. Three chamber designs were produced to determine the influence of the ion chamber response on electrode separation, field strength, and collection volume on the ion chamber response under UHDR and ultra-high dose per pulse (UHDPP) conditions. METHODS Three chambers were designed and produced: the A11-VAR (0.2-1.0 mm electrode gap, 20 mm diameter collector), the A11-TPP (0.3 mm electrode gap, 20 mm diameter collector), and the A30 (0.3 mm electrode gap, 5.4 mm diameter collector). The chambers underwent full characterization using an UHDR 9 MeV electron beam with individually varied beam parameters of pulse repetition frequency (PRF, 10-120 Hz), pulse width (PW, 0.5-4 µs), and pulse amplitude (0.01-9 Gy/pulse). The response of the ion chambers was evaluated as a function of the DPP, PRF, PW, dose rate, electric field strength, and electrode gap. RESULTS The chamber response was found to be dependent on DPP and PW, and these dependencies were mitigated with larger electric field strengths and smaller electrode spacing. At a constant electric field strength, we measured a larger CCE as a function of DPP for ion chambers with a smaller electrode gap in the A11-VAR. For ion chambers with identical electrode gap (A11-TPP and A30), higher electric field strengths were found to yield better CCE at higher DPP. A PW dependence was observed at low electric field strengths (500 V/mm) for DPP values ranging from 1 to 5 Gy at PWs ranging from 0.5 to 4 µs, but at electric field strengths of 1000 V/mm and higher, these effects become negligible. CONCLUSION This study confirmed that the CCE of ion chambers depends strongly on the electrode spacing and the electric field strength, and also on the DPP and the PW of the UHDR beam. A significant finding of this study is that although chamber performance does depend on PW, the effect on the CCE becomes negligible with reduced electrode spacing and increased electric field. A CCE of ≥95% was achieved for DPPs of up to 5 Gy with no observable dependence on PW using the A30 chamber, while still achieving an acceptable performance in conventional dose rate beams, opening up the possibility for this type of chamber to be used as a reference class chamber for calibration purposes of electron FLASH beamlines.
Collapse
Affiliation(s)
- Kevin Liu
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, Texas, USA
| | | | - Ahtesham Ullah Khan
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Radiation Oncology, Northwestern Memorial Hospital, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Brian Hooten
- Standard Imaging Inc., Middleton, Wisconsin, USA
| | - Larry DeWerd
- Department of Medical Physics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Emil Schüler
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Sam Beddar
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, Texas, USA
| |
Collapse
|
19
|
Li M, Zhou S, Dong G, Wang C. Emergence of FLASH‑radiotherapy across the last 50 years (Review). Oncol Lett 2024; 28:602. [PMID: 39493433 PMCID: PMC11529378 DOI: 10.3892/ol.2024.14735] [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: 05/16/2024] [Accepted: 09/02/2024] [Indexed: 11/05/2024] Open
Abstract
A novel radiotherapy (RT) approach termed FLASH-RT, which irradiates areas at ultra-high dose rates, is of current interest to medical researchers. FLASH-RT can maintain equivalent antitumor effects while sparing healthy tissue compared with conventional RT (CONV-RT), which uses low dose rates. The sparing effect on healthy tissue after FLASH-RT is known as the FLASH effect. Owing to the FLASH effect, FLASH-RT can raise the maximum tolerable dose to control tumor growth or eradicate the tumor and provide a new strategy for clinical RT. However, definitive irradiation conditions for reproducing the FLASH effect and the biological mechanism of the FLASH effect have not yet been fully elucidated. The efficacy of FLASH-RT is controversial despite its successful application in clinical RT. The present review recapitulates the progression of FLASH-RT and critically comments on the hypothesis of the FLASH effect. In addition, the review expounds on the current issues with regard to the differential phenomena between in vitro and in vivo studies, and elaborates on the challenges for the application of FLASH-RT that need to be addressed in the future.
Collapse
Affiliation(s)
- Menghua Li
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
- Department of Biochemistry, Basic Medical College of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Sen Zhou
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Guofu Dong
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
- Department of Biochemistry, Basic Medical College of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Changzhen Wang
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
- Department of Biochemistry, Basic Medical College of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
20
|
Saini J, Erickson DPJ, Vander Stappen F, Ruth M, Cui S, Gorman V, Rossomme S, Cao N, Ford EC, Meyer J, Bloch C, Wong T, Grassberger C, Rengan R, Zeng J, Schwarz M. Commissioning a clinical proton pencil beam scanning beamline for pre-clinical ultra-high dose rate irradiations on a cyclotron-based system. Front Oncol 2024; 14:1460288. [PMID: 39678495 PMCID: PMC11638781 DOI: 10.3389/fonc.2024.1460288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/22/2024] [Indexed: 12/17/2024] Open
Abstract
Background This manuscript describes modifications to a pencil beam scanning (PBS) proton gantry that enables ultra-high dose rates (UHDR) irradiation, including treatment planning and validation. Methods Beamline modifications consisted of opening the energy slits and setting the degrader to pass-through mode to maximize the dose rate. A range shifter was inserted upstream from the isocenter to enlarge the spot size and make it rotationally symmetric. We measured the beamline transport efficiency and investigated the variation in output due to the recombination of charge in the dose monitoring chamber. The output calibration was performed through a parallel plate chamber (PPC05), and an intercomparison was performed for various detectors. The pre-clinical field for mice irradiation consisted of different dose levels to deliver uniform doses in transmission mode. The field dose rates were determined through log files while scripting in TPS was used to estimate PBS dose rates. The survival experiments consisted of irradiating the full pelvis of the mice at UHDR and conventional dose rates. Results The spot size was constant with beam current and had a sigma of 8.5 mm at the isocenter. The beam output increased by 35% at 720 nA compared to 5.6 nA, primarily due to recombination in the dose-monitoring ion chambers. The Faraday Cup and PPC05 agreed within 2%, while other detectors were within 3% of FC for dose rates <60 Gy/s. The pre-clinical fields' PBS dose rate is above 45 Gy/sec for all voxels within the target volume. The average and PBS dose rates decrease as field size increases and approaches 40 Gy/s for a field size of 7x7 cm2. All UHDR arms showed better survival than the corresponding conventional dose rate arms. Conclusions We successfully modified a clinical system to perform UHDR pre-clinical experiments. As part of our pre-clinical experiments, we observed the FLASH effect concerning mice survival.
Collapse
Affiliation(s)
- Jatinder Saini
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, WA, United States
- Radiation Oncology, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | | | | | - Matt Ruth
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, WA, United States
- Radiation Oncology, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Sunan Cui
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, WA, United States
- Radiation Oncology, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Vanessa Gorman
- Proton Therapy - Research and Development, Ion Beam Applications, Louvain-La-Neuve, Belgium
| | - Séverine Rossomme
- Proton Therapy - Research and Development, Ion Beam Applications, Louvain-La-Neuve, Belgium
| | - Ning Cao
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, WA, United States
- Radiation Oncology, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Eric C. Ford
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, WA, United States
- Radiation Oncology, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Juergen Meyer
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, WA, United States
- Radiation Oncology, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Charles Bloch
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, WA, United States
- Radiation Oncology, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Tony Wong
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, WA, United States
- Radiation Oncology, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Clemens Grassberger
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, WA, United States
| | - Ramesh Rengan
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, WA, United States
- Radiation Oncology, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Jing Zeng
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, WA, United States
- Radiation Oncology, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Marco Schwarz
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, WA, United States
- Radiation Oncology, Fred Hutchinson Cancer Center, Seattle, WA, United States
| |
Collapse
|
21
|
Alhaddad L, Osipov AN, Leonov S. FLASH Radiotherapy: Benefits, Mechanisms, and Obstacles to Its Clinical Application. Int J Mol Sci 2024; 25:12506. [PMID: 39684218 DOI: 10.3390/ijms252312506] [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: 10/08/2024] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 12/18/2024] Open
Abstract
Radiotherapy (RT) has been shown to be a cornerstone of both palliative and curative tumor care. RT has generally been reported to be sharply limited by ionizing radiation (IR)-induced toxicity, thereby constraining the control effect of RT on tumor growth. FLASH-RT is the delivery of ultra-high dose rate (UHDR) several orders of magnitude higher than what is presently used in conventional RT (CONV-RT). The FLASH-RT clinical trials have been designed to examine the UHDR deliverability, the effectiveness of tumor control, the dose tolerance of normal tissue, and the reproducibility of treatment effects across several institutions. Although it is still in its infancy, FLASH-RT has been shown to have potential to rival current RT in terms of safety. Several studies have suggested that the adoption of FLASH-RT is very limited, and the incorporation of this new technique into routine clinical RT will require the use of accurate dosimetry methods and reproducible equipment that enable the reliable and robust measurements of doses and dose rates. The purpose of this review is to highlight the advantages of this technology, the potential mechanisms underpinning the FLASH-RT effect, and the major challenges that need to be tackled in the clinical transfer of FLASH-RT.
Collapse
Affiliation(s)
- Lina Alhaddad
- Department of Environmental Sciences, Faculty of Science, Damascus University, Damascus P.O. Box 30621, Syria
| | - Andreyan N Osipov
- N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow 119991, Russia
- Moscow Center for Advanced Studies, Kulakova Str. 20, Moscow 123592, Russia
- State Research Center-Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency (SRC-FMBC), Moscow 123098, Russia
- CANDLE Synchrotron Research Institute, 31 Acharyan, Yerevan 0040, Armenia
| | - Sergey Leonov
- Moscow Center for Advanced Studies, Kulakova Str. 20, Moscow 123592, Russia
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia
| |
Collapse
|
22
|
Zhao X, Huang S, Lin H, Choi JI, Zhu K, Simone CB, Yan X, Kang M. A Novel Dose Rate Optimization Method to Maximize Ultrahigh-Dose-Rate Coverage of Critical Organs at Risk Without Compromising Dosimetry Metrics in Proton Pencil Beam Scanning FLASH Radiation Therapy. Int J Radiat Oncol Biol Phys 2024; 120:1181-1191. [PMID: 38879087 DOI: 10.1016/j.ijrobp.2024.06.002] [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: 03/01/2023] [Revised: 05/12/2024] [Accepted: 06/09/2024] [Indexed: 07/10/2024]
Abstract
PURPOSE This study aimed to investigate a dose rate optimization framework based on the spot-scanning patterns to improve ultrahigh-dose-rate coverage of critical organs at risk (OARs) for proton pencil beam scanning (PBS) FLASH radiation therapy (ultrahigh dose-rate (often referred to as >40 Gy per second) delivery) and present implementation of a genetic algorithm (GA) method for spot sequence optimization to achieve PBS FLASH dose rate optimization under relatively low nozzle beam currents. METHODS AND MATERIALS First, a multifield FLASH plan was developed to meet all the dosimetric goals and optimal FLASH dose rate coverage by considering the deliverable minimum monitor unit constraint. Then, a GA method was implemented into the in-house treatment platform to maximize the dose rate by exploring the best spot delivery sequence. A phantom study was performed to evaluate the effectiveness of the dose rate optimization. Then, 10 consecutive plans for patients with lung cancer previously treated using PBS intensity-modulated proton therapy were optimized using 45 GyRBE in 3 fractions for both transmission and Bragg peak FLASH radiation therapy for further validation. The spot delivery sequence of each treatment field was optimized using this GA. The ultrahigh-dose-rate-volume histogram and dose rate coverage V40GyRBE/s were investigated to assess the efficacy of dose rate optimization quantitatively. RESULTS Using a relatively low monitor unit/spot of 150, corresponding to a nozzle beam current of 65 nA, the FLASH dose rate ratio V40GyRBE/s of the OAR contour of the core was increased from 0% to ∼60% in the phantom study. In the patients with lung cancer, the ultrahigh-dose-rate coverage V40GyRBE/s was improved from 15.2%, 15.5%, 17.6%, and 16.0% before the delivery sequence optimization to 31.8%, 43.5%, 47.6%, and 30.5% after delivery sequence optimization in the lungs-GTV (gross tumor volume), spinal cord, esophagus, and heart (for all, P < .001). When the beam current increased to 130 nA, V40GyRBE/s was improved from 45.1%, 47.1%, 51.2%, and 51.4% to 65.3%, 83.5%, 88.1%, and 69.4% (P < .05). The averaged V40GyRBE/s for the target and OARs increased from 12.9% to 41.6% and 46.3% to 77.5% for 65 and 130 nA, respectively, showing significant improvements based on a clinical proton system. After optimizing the dose rate for the Bragg peak FLASH technique with a beam current of 340 nA, the V40GyRBE/s values for the lung GTV, spinal cord, esophagus, and heart were increased by 8.9%, 15.8%, 22%, and 20.8%, respectively. CONCLUSIONS An optimal plan quality can be maintained as the spot delivery sequence optimization is a separate independent process after the plan optimization. Both the phantom and patient results demonstrated that novel spot delivery sequence optimization can effectively improve the ultrahigh-dose-rate coverage for critical OARs, which can potentially be applied in clinical practice for better OARs-sparing efficacy.
Collapse
Affiliation(s)
- Xingyi Zhao
- State Key Laboratory of Nuclear Physics and Technology, and Key Laboratory of HEDP of the Ministry of Education, Center for Applied Physics and Technology, Peking University, Beijing, China; New York Proton Center, New York, New York
| | - Sheng Huang
- Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Haibo Lin
- New York Proton Center, New York, New York; Departments of Radiation Oncology and Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - J Isabelle Choi
- New York Proton Center, New York, New York; Departments of Radiation Oncology and Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kun Zhu
- State Key Laboratory of Nuclear Physics and Technology, and Key Laboratory of HEDP of the Ministry of Education, Center for Applied Physics and Technology, Peking University, Beijing, China
| | - Charles B Simone
- New York Proton Center, New York, New York; Departments of Radiation Oncology and Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Xueqing Yan
- State Key Laboratory of Nuclear Physics and Technology, and Key Laboratory of HEDP of the Ministry of Education, Center for Applied Physics and Technology, Peking University, Beijing, China.
| | - Minglei Kang
- New York Proton Center, New York, New York; Departments of Radiation Oncology and Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
23
|
Scarmelotto A, Delprat V, Michiels C, Lucas S, Heuskin AC. The oxygen puzzle in FLASH radiotherapy: A comprehensive review and experimental outlook. Clin Transl Radiat Oncol 2024; 49:100860. [PMID: 39381632 PMCID: PMC11458961 DOI: 10.1016/j.ctro.2024.100860] [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: 05/21/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 10/10/2024] Open
Abstract
FLASH radiotherapy is attracting increasing interest because it maintains tumor control while inflicting less damage to normal tissues compared to conventional radiotherapy. This sparing effect, the so-called FLASH effect, is achieved when radiation is delivered at ultra-high dose rates (≥40 Gy/s). Although the FLASH effect has already been demonstrated in several preclinical models, a complete mechanistic description explaining why tumors and normal tissues respond differently is still missing. None of the current hypotheses fully explains the experimental evidence. A common point between many of these is the role of oxygen, which is described as a major factor, either through transient hypoxia in the form of dissolved molecules, or reactive oxygen species (ROS). Therefore, this review focuses on both forms of this molecule, retracing old and more recent theories, while proposing new mechanisms that could provide a complete description of the FLASH effect based on preclinical and experimental evidence. In addition, this manuscript describes a set of experiments designed to provide the FLASH community with new tools for exploring the post-irradiation fate of ROS and their potential biological implications.
Collapse
Affiliation(s)
- Andrea Scarmelotto
- Laboratory for Analysis by Nuclear Reaction (LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Victor Delprat
- Laboratory for Analysis by Nuclear Reaction (LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Carine Michiels
- Unité de Recherche en Biologie Cellulaire (URBC), Namur Research Institute For Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
| | - Stéphane Lucas
- Laboratory for Analysis by Nuclear Reaction (LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
- Ion Beam Application (IBA), Chemin du Cyclotron, 6, B-1348 Louvain-La-Neuve, Belgium
| | - Anne-Catherine Heuskin
- Laboratory for Analysis by Nuclear Reaction (LARN), Namur Research Institute for Life Sciences (NARILIS), University of Namur, Rue de Bruxelles 61, B-5000 Namur, Belgium
| |
Collapse
|
24
|
Tobias Böhlen T, Psoroulas S, Aylward JD, Beddar S, Douralis A, Delpon G, Garibaldi C, Gasparini A, Schüler E, Stephan F, Moeckli R, Subiel A. Recording and reporting of ultra-high dose rate "FLASH" delivery for preclinical and clinical settings. Radiother Oncol 2024; 200:110507. [PMID: 39245070 DOI: 10.1016/j.radonc.2024.110507] [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: 01/26/2024] [Revised: 08/08/2024] [Accepted: 08/19/2024] [Indexed: 09/10/2024]
Abstract
Treatments at ultra-high dose rate (UHDR) have the potential to improve the therapeutic index of radiation therapy (RT) by sparing normal tissues compared to conventional dose rate irradiations. Insufficient and inconsistent reporting in physics and dosimetry of preclinical and translational studies may have contributed to a reproducibility crisis of radiobiological data in the field. Consequently, the development of a common terminology, as well as common recording, reporting, dosimetry, and metrology standards is required. In the context of UHDR irradiations, the temporal dose delivery parameters are of importance, and under-reporting of these parameters is also a concern.This work proposes a standardization of terminology, recording, and reporting to enhance comparability of both preclinical and clinical UHDR studies and and to allow retrospective analyses to aid the understanding of the conditions which give rise to the FLASH effect.
Collapse
Affiliation(s)
- Till Tobias Böhlen
- Institute of Radiation Physics, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Serena Psoroulas
- Center for Proton Therapy, Paul Scherrer Institute, Villigen PSI, Switzerland; Klinik für Radio-Onkologie, UniversitätsSpital Zürich, Switzerland
| | - Jack D Aylward
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK; Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, UK; Medical Physics, School of Applied Sciences, University of the West of England, Bristol, UK
| | - Sam Beddar
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Grégory Delpon
- Institut de Cancérologie de l'Ouest, Medical Physics Department, Saint-Herblain, France; Nantes Université, IMT Atlantique, CNRS/IN2P3, SUBATECH, Nantes, France
| | - Cristina Garibaldi
- IEO, Unit of Radiation Research, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Alessia Gasparini
- CORE, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Medical Physics Department, Iridium Netwerk, Wilrijk, Belgium
| | - Emil Schüler
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Frank Stephan
- Deutsches Elektronen-Synchrotron DESY, Platanenallee 6, 15738 Zeuthen, Germany
| | - Raphaël Moeckli
- Institute of Radiation Physics, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland.
| | - Anna Subiel
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, UK; University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
25
|
Pogue BW, Thomas WS, Tavakkoli AD, Jarvis LA, Hoopes PJ. Major contributors to FLASH sparing efficacy emerge from murine skin studies: dose rate, total dose per fraction, anesthesia and oxygenation. Front Oncol 2024; 14:1414584. [PMID: 39525619 PMCID: PMC11544430 DOI: 10.3389/fonc.2024.1414584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024] Open
Abstract
Background Normal tissue sparing from radiation damage upon ultra-high dose rate irradiation, known as the FLASH effect with an equivalent tumor response, has been widely reported in murine skin models, and translation of this type of radiotherapy to humans has already begun, with skin sparing being a primary outcome expected. Methods This study reviews the status of the field, focusing on the proposed mechanisms and skin response assays, outlining what has become known in terms of input parameters that might control the magnitude of the FLASH effect. Results Murine studies have largely focused on acute damage responses, developing over 3-8 weeks, to single doses of FLASH versus conventional dose rate (CDR), suggesting that at dose rates above tens of Gray per second, with a total dose of more than 20 Gy, the FLASH effect is induced. Fractionated delivery appears to be possible, although fraction sizes >17 Gy appear to be needed for sparing efficacy. The interplay between the dose rate and total dose per fraction remains to be fully elucidated. Oxygen is a modulator of efficacy, with both hypoxia and hyperoxia diminishing the effect of FLASH. Measurement of transient changes in oxygen levels is possible and may be a marker of treatment efficacy. Conclusion Taken together, murine skin data provide important information for translational studies, despite the associated limitations. Studies of later-term sparing effects, as well as studies on pig skin, are needed to take the next step in assessing translational FLASH efficacy. The control of biological factors, such as tissue oxygenation, may be required to understand and control the response.
Collapse
Affiliation(s)
- Brian W. Pogue
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, United States
- Thayer School of Engineering at Dartmouth, Hanover, NH, United States
| | - William S. Thomas
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, United States
| | - Armin D. Tavakkoli
- Department of Surgery, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Lesley A. Jarvis
- Thayer School of Engineering at Dartmouth, Hanover, NH, United States
- Department of Surgery, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - P. Jack Hoopes
- Thayer School of Engineering at Dartmouth, Hanover, NH, United States
- Department of Surgery, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| |
Collapse
|
26
|
González-Crespo I, Gómez F, López Pouso Ó, Pardo-Montero J. An in-silico study of conventional and FLASH radiotherapy iso-effectiveness: potential impact of radiolytic oxygen depletion on tumor growth curves and tumor control probability. Phys Med Biol 2024; 69:215016. [PMID: 39357538 DOI: 10.1088/1361-6560/ad8291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 10/01/2024] [Indexed: 10/04/2024]
Abstract
Objective. This work aims to investigate the iso-effectiveness of conventional and FLASH radiotherapy on tumors through in-silico mathematical models. We focused on the role of radiolytic oxygen depletion (ROD), which has been argued as a possible factor to explain the FLASH effect.Approach. We used a spatiotemporal reaction-diffusion model, including ROD, to simulate tumor oxygenation and response. From those oxygen distributions we obtained surviving fractions (SFs) using the linear-quadratic (LQ) model with the oxygen enhancement ratios (OERs). We then employed the calculated SFs to describe the evolution of preclinical tumor volumes through a mathematical model of tumor response, and we also extrapolated those results to calculate tumor control probabilities (TCPs) using the Poisson-LQ approach.Main results. Our study suggests that the ROD effect may cause differences in SF between FLASH and conventional radiotherapy, especially in lowα/βandpoorly oxygenatedcells. However, a statistical analysis showed that these changes in SF generally do not result in significant differences in the evolution of preclinical tumor growth curves when the sample size is small, because such differences in SF may not be noticeable in the heterogeneity of the population of animals. Nonetheless, when extrapolating this effect to TCP curves, we observed important differences between both techniques (TCP is lower in FLASH radiotherapy). When analyzing the response of tumors with heterogeneous oxygenations, differences in TCP are more important forwell oxygenatedtumors. This apparent contradiction with the results obtained for homogeneously oxygenated cells is explained by the complex interplay between the heterogeneity of tumor oxygenation, the OER effect, and the ROD effect.Significance. This study supports the experimentally observed iso-effectiveness of FLASH and conventional radiotherapy when analyzing the volume evolution of preclinical tumors (that are far from control). However, this study also hints that tumor growth curves may be less sensitive to small variations in SF than tumor control probability: ROD may lead to increased SF in FLASH radiotherapy, which while not large enough to cause significant differences in tumor growth curves, could lead to important differences in clinical TCPs. Nonetheless, it cannot be discarded that other effects not modeled in this work, like radiation-induced immune effects, can contribute to tumor control and maintain the iso-effectiveness of FLASH radiotherapy. The study of tumor growth curves may not be the ideal experiment to test the iso-effectiveness of FLASH, and experiments reporting TCP orD50may be preferred.
Collapse
Affiliation(s)
- I González-Crespo
- Group of Medical Physics and Biomathematics, Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
- Department of Applied Mathematics, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - F Gómez
- Department of Particle Physics, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Ó López Pouso
- Group of Medical Physics and Biomathematics, Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
- Department of Applied Mathematics, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Galician Centre for Mathematical Research and Technology (CITMAga), Santiago de Compostela, Spain
| | - J Pardo-Montero
- Group of Medical Physics and Biomathematics, Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
- Department of Medical Physics, Complexo Hospitalario Universitario de Santiago, Santiago de Compostela, Spain
| |
Collapse
|
27
|
Liu K, Waldrop T, Aguilar E, Mims N, Neill D, Delahoussaye A, Li Z, Swanson D, Lin SH, Koong AC, Taniguchi CM, Loo BW, Mitra D, Schüler E. Redefining FLASH Radiation Therapy: The Impact of Mean Dose Rate and Dose Per Pulse in the Gastrointestinal Tract. Int J Radiat Oncol Biol Phys 2024:S0360-3016(24)03466-7. [PMID: 39424078 DOI: 10.1016/j.ijrobp.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/07/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024]
Abstract
PURPOSE The understanding of how varying radiation beam parameter settings affect the induction and magnitude of the FLASH effect remains limited. We sought to systematically evaluate how the magnitude of radiation-induced gastrointestinal toxicity depends on the interplay between mean dose rate (MDR) and dose per pulse (DPP). METHODS AND MATERIALS C57BL/6J mice received total abdominal irradiation (TAI, 11-14 Gy single fraction) through either conventional (CONV) irradiation (low-DPP and low MDR, CONV) or through various combinations of DPP and MDR up to ultra-high-dose-rate beam conditions. DPPs ranging from 1 to 6 Gy were evaluated, while the total dose and MDR (>100 Gy/s) were kept constant; the effects of MDR were evaluated for the range of 0.3 to 1440 Gy/s, while the total dose and DPP were kept constant. Radiation-induced gastrointestinal toxicity was quantified in nontumor-bearing mice through the regenerating crypt assay and survival assessment. Tumor response was evaluated through tumor growth delay. RESULTS Within each tested total dose using a constant MDR (>100 Gy/s), increasing DPP led to an increase in sparing (an increase in the number of regenerating crypts), with a more prominent effect seen at 12- and 14-Gy TAI. Interestingly, at DPPs of >4 Gy, a similar level of crypt sparing was demonstrated irrespective of the MDR used (from 0.3 to 1440 Gy/s). At a fixed high-DPP of 4.7 Gy, survival was equivalently improved relative to CONV irrespective of MDR. However, at a lower DPP of 0.93 Gy, an MDR of 104 Gy/s produced a greater survival effect compared with 0.3 Gy/s. We also confirmed that high-DPP, regardless of MDR, produced the same magnitude of tumor growth delay relative to CONV using a clinically relevant melanoma mouse model. CONCLUSIONS This study demonstrates the strong influence that the beam parameter settings have on the magnitude of the FLASH effect. Both high-DPP and ultra-high-dose-rate appeared independently sufficient to produce FLASH sparing of gastrointestinal toxicity while isoeffective tumor response was maintained across all conditions.
Collapse
Affiliation(s)
- Kevin Liu
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas; The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Trey Waldrop
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas; The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Edgardo Aguilar
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nefetiti Mims
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Denae Neill
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Abagail Delahoussaye
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ziyi Li
- Division of Basic Sciences, Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David Swanson
- Division of Basic Sciences, Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Steven H Lin
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Albert C Koong
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cullen M Taniguchi
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Billy W Loo
- Department of Radiation Oncology & Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Devarati Mitra
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Emil Schüler
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas; The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, Texas.
| |
Collapse
|
28
|
Simeonov Y, Weber U, Krieger M, Schuy C, Folkerts M, Paquet G, Lansonneur P, Penchev P, Zink K. A Fast 3D Range-Modulator Delivery Approach: Validation of the FLUKA Model on a Varian ProBeam System Including a Robustness Analysis. Cancers (Basel) 2024; 16:3498. [PMID: 39456592 PMCID: PMC11505765 DOI: 10.3390/cancers16203498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/09/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
A 3D range-modulator (RM), optimized for a single energy and a specific target shape, is a promising and viable solution for the ultra-fast dose delivery in particle therapy. The aim of this work was to investigate the impact of potential beam and modulator misalignments on the dose distribution. Moreover, the FLUKA Monte Carlo model, capable of simulating 3D RMs, was adjusted and validated for the 250 MeV single-energy proton irradiation from a Varian ProBeam system. A 3D RM was designed for a cube target shape rotated 45° around two axes using a Varian-internal research version of the Eclipse treatment planning software, and the resulting dose distribution was simulated in a water phantom. Deviations from the ideal alignment were introduced, and the dose distributions from the modified simulations were compared to the original unmodified one. Finally, the FLUKA model and the workflow were validated with base-line data measurements and dose measurements of the manufactured modulator prototype at the HollandPTC facility in Delft. The adjusted FLUKA model, optimized particularly in the scope of a single-energy FLASH irradiation with a PMMA pre-absorber, demonstrated very good agreement with the measured dose distribution resulting from the 3D RM. Dose deviations resulting from modulator-beam axis misalignments depend on the specific 3D RM and its shape, pin aspect ratio, rotation angle, rotation point, etc. A minor modulator shift was found to be more relevant for the distal dose distribution than for the spread-out Bragg Peak (SOBP) homogeneity. On the other hand, a modulator tilt (rotation away from the beam axis) substantially affected not only the depth dose profile, transforming a flat SOBP into a broad, Gaussian-like distribution with increasing rotation angle, but also shifted the lateral dose distribution considerably. This work strives to increase awareness and highlight potential pitfalls as the 3D RM method progresses from a purely research concept to pre-clinical studies and human trials. Ensuring that gantry rotation and the combined weight of RM, PMMA, and aperture do not introduce alignment issues is critical. Given all the other range and positioning uncertainties, etc., not related to the modulator, the RM must be aligned with an accuracy below 1° in order to preserve a clinically acceptable total uncertainty budget. Careful consideration of critical parameters like the pin aspect ratio and possibly a novel robust modulator geometry optimization are potential additional strategies to mitigate the impact of positioning on the resulting dose. Finally, even the rotated cube 3D modulator with high aspect ratio pin structures (~80 mm height to 3 mm pin base width) was found to be relatively robust against a slight misalignment of 0.5° rotation or a 1.5 mm shift in one dimension perpendicular to the beam axis. Given a reliable positioning and QA concept, the additional uncertainties introduced by the 3D RM can be successfully managed adopting the concept into the clinical routine.
Collapse
Affiliation(s)
- Yuri Simeonov
- Institute of Medical Physics and Radiation Protection (IMPS), University of Applied Sciences, 35390 Giessen, Germany; (U.W.); (P.P.); (K.Z.)
| | - Ulrich Weber
- Institute of Medical Physics and Radiation Protection (IMPS), University of Applied Sciences, 35390 Giessen, Germany; (U.W.); (P.P.); (K.Z.)
- Biophysics Division, GSI Helmholtzzentrum für Schwerionenforschung GmbH, 64291 Darmstadt, Germany;
| | - Miriam Krieger
- Varian Medical Systems, Palo Alto, CA 94304, USA; (M.K.); (M.F.); (G.P.)
| | - Christoph Schuy
- Biophysics Division, GSI Helmholtzzentrum für Schwerionenforschung GmbH, 64291 Darmstadt, Germany;
| | - Michael Folkerts
- Varian Medical Systems, Palo Alto, CA 94304, USA; (M.K.); (M.F.); (G.P.)
| | - Gerard Paquet
- Varian Medical Systems, Palo Alto, CA 94304, USA; (M.K.); (M.F.); (G.P.)
| | - Pierre Lansonneur
- Varian Medical Systems, Palo Alto, CA 94304, USA; (M.K.); (M.F.); (G.P.)
| | - Petar Penchev
- Institute of Medical Physics and Radiation Protection (IMPS), University of Applied Sciences, 35390 Giessen, Germany; (U.W.); (P.P.); (K.Z.)
| | - Klemens Zink
- Institute of Medical Physics and Radiation Protection (IMPS), University of Applied Sciences, 35390 Giessen, Germany; (U.W.); (P.P.); (K.Z.)
- Marburg Ion Beam Therapy Center (MIT), 35043 Marburg, Germany
| |
Collapse
|
29
|
Ma Y, Zhang W, Zhao Z, Lv J, Chen J, Yan X, Lin X, Zhang J, Wang B, Gao S, Xiao J, Yang G. Current views on mechanisms of the FLASH effect in cancer radiotherapy. Natl Sci Rev 2024; 11:nwae350. [PMID: 39479528 PMCID: PMC11523052 DOI: 10.1093/nsr/nwae350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 11/02/2024] Open
Abstract
FLASH radiotherapy (FLASH-RT) is a new modality of radiotherapy that delivers doses with ultra-high dose rates. The FLASH effect was defined as the ability of FLASH-RT to suppress tumor growth while sparing normal tissues. Although the FLASH effect has been proven to be valid in various models by different modalities of irradiation and clinical trials of FLASH-RT have achieved promising initial success, the exact underlying mechanism is still unclear. This article summarizes mainstream hypotheses of the FLASH effect at physicochemical and biological levels, including oxygen depletion and free radical reactions, nuclear and mitochondria damage, as well as immune response. These hypotheses contribute reasonable explanations to the FLASH effect and are interconnected according to the chronological order of the organism's response to ionizing radiation. By collating the existing consensus, evidence and hypotheses, this article provides a comprehensive overview of potential mechanisms of the FLASH effect and practical guidance for future investigation in the field of FLASH-RT.
Collapse
Affiliation(s)
- Yuqi Ma
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China
| | - Wenkang Zhang
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China
| | - Ziming Zhao
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China
| | - Jianfeng Lv
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China
| | - Junyi Chen
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China
| | - Xueqin Yan
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China
| | - XiaoJi Lin
- Oncology Discipline Group, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325003, China
| | - Junlong Zhang
- Beijing National Laboratory of Molecular Science, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Bingwu Wang
- Beijing National Laboratory of Molecular Science, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Song Gao
- Beijing National Laboratory of Molecular Science, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, China
| | - Jie Xiao
- KIRI Precision Particle Therapy Flash Technologies Research Center, Guangzhou 510700, China
| | - Gen Yang
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, Peking University, Beijing 100871, China
| |
Collapse
|
30
|
Chaikh A, Édouard M, Huet C, Milliat F, Villagrasa C, Isambert A. Towards clinical application of ultra-high dose rate radiotherapy and the FLASH effect: Challenges and current status. Cancer Radiother 2024; 28:463-473. [PMID: 39304401 DOI: 10.1016/j.canrad.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 09/22/2024]
Abstract
Ultra-high dose rate external beam radiotherapy (UHDR-RT) uses dose rates of several tens to thousands of Gy/s, compared with the dose rate of the order of a few Gy/min for conventional radiotherapy techniques, currently used in clinical practice. The use of such dose rate is likely to improve the therapeutic index by obtaining a radiobiological effect, known as the "FLASH" effect. This would maintain tumor control while enhancing tissues protection. To date, this effect has been achieved using beams of electrons, photons, protons, and heavy ions. However, the conditions required to achieve this "FLASH" effect are not well defined, and raise several questions, particularly with regard to the definition of the prescription, including dose fractionation, irradiated volume and the temporal structure of the pulsed beam. In addition, the dose delivered over a very short period induces technical challenges, particularly in terms of detectors, which must be mastered to guarantee safe clinical implementation. IRSN has carried out an in-depth literature review of the UHDR-RT technique, covering various aspects relating to patient radiation protection: the radiobiological mechanisms associated with the FLASH effect, the used temporal structure of the UHDR beams, accelerators and dose control, the properties of detectors to be used with UHDR beams, planning, clinical implementation, and clinical studies already carried out or in progress.
Collapse
Affiliation(s)
| | | | | | - Fabien Milliat
- IRSN/PSE-SANTÉ-SERAMED/LRMed, Fontenay-aux-Roses, France
| | | | | |
Collapse
|
31
|
Gesualdi F, de Marzi L, Dutreix M, Favaudon V, Fouillade C, Heinrich S. A multidisciplinary view of flash irradiation. Cancer Radiother 2024; 28:453-462. [PMID: 39343695 DOI: 10.1016/j.canrad.2024.07.003] [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: 04/15/2024] [Revised: 07/19/2024] [Accepted: 07/19/2024] [Indexed: 10/01/2024]
Abstract
The delivery of ultra-high dose rates of radiation, called flash irradiation or flash-RT, has emerged as a new modality of radiotherapy shaking up the paradigm of proportionality of effect and dose whatever the method of delivery of the radiation. The hallmark of flash-RT is healthy tissue sparing from the side effects of radiation without decrease of the antitumor efficiency in animal models. In this review we will define its specificities, the molecular mechanisms underlying the flash effect and the ongoing developments to bring this new modality to patient treatment.
Collapse
Affiliation(s)
- Flavia Gesualdi
- Institut Curie, Hospital Division, centre de protonthérapie d'Orsay, université Paris-Saclay, université PSL, centre universitaire, 91948 Orsay cedex, France
| | - Ludovic de Marzi
- Institut Curie, Hospital Division, centre de protonthérapie d'Orsay, université Paris-Saclay, université PSL, centre universitaire, 91948 Orsay cedex, France; Institut Curie, université PSL, université Paris-Saclay, Inserm Lito U1288, centre universitaire, 91898 Orsay, France
| | - Marie Dutreix
- Institut Curie, Research Division, Inserm U 1021-CNRS UMR 3347, université Paris-Saclay, université PSL, centre universitaire, 91401 Orsay cedex, France
| | - Vincent Favaudon
- Institut Curie, Research Division, Inserm U 1021-CNRS UMR 3347, université Paris-Saclay, université PSL, centre universitaire, 91401 Orsay cedex, France
| | - Charles Fouillade
- Institut Curie, Research Division, Inserm U 1021-CNRS UMR 3347, université Paris-Saclay, université PSL, centre universitaire, 91401 Orsay cedex, France
| | - Sophie Heinrich
- Institut Curie, Research Division, Inserm U 1021-CNRS UMR 3347, université Paris-Saclay, université PSL, centre universitaire, 91401 Orsay cedex, France.
| |
Collapse
|
32
|
Cheng C, Xu L, Jing H, Selvaraj B, Lin H, Pennock M, Chhabra AM, Hasan S, Zhai H, Zhang Y, Nie K, Bakst RL, Kabarriti R, Choi JI, Lee NY, Simone CB, Kang M, Wu H. The Potential and Challenges of Proton FLASH in Head and Neck Cancer Reirradiation. Cancers (Basel) 2024; 16:3249. [PMID: 39409872 PMCID: PMC11482542 DOI: 10.3390/cancers16193249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024] Open
Abstract
Ultrahigh-dose-rate therapy, also known as FLASH radiotherapy (RT), is an emerging technique that is garnering significant interest in cancer treatment due to its potential to revolutionize therapy. This method can achieve comparable tumor control to conventional-dose-rate RT while offering the enhanced protection of normal tissue through the FLASH-sparing effect. This innovative technique has demonstrated promising results in preclinical studies involving animals and cell lines. Particularly noteworthy is its potential application in treating head and neck (HN) cancers, especially in patients with challenging recurrent tumors and reirradiation cases, where the toxicity rates with conventional radiotherapy are high. Such applications aim to enhance tumor control while minimizing side effects and preserving patients' quality of life. In comparison to electron or photon FLASH modalities, proton therapy has demonstrated superior dosimetric and delivery characteristics and is a safe and effective FLASH treatment for human malignancies. Compared to the transmission proton FLASH, single-energy Bragg peak FLASH is a novel delivery method that allows highly conformal doses to targets and minimal radiation doses to crucial OARs. Proton Bragg peak FLASH for HN cancer has still not been well studied. This review highlights the significance of proton FLASH in enhancing cancer therapy by examining the advantages and challenges of using it for HN cancer reirradiation.
Collapse
Affiliation(s)
- Chingyun Cheng
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA; (C.C.)
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Liming Xu
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Hao Jing
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | | | - Haibo Lin
- New York Proton Center, New York, NY 10035, USA
| | - Michael Pennock
- Department of Radiation Oncology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA
| | | | | | | | - Yin Zhang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA; (C.C.)
| | - Ke Nie
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA; (C.C.)
| | - Richard L. Bakst
- Department of Radiation Oncology–Radiation Oncology Associates, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rafi Kabarriti
- Department of Radiation Oncology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10461, USA
| | - J. Isabelle Choi
- New York Proton Center, New York, NY 10035, USA
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Nancy Y. Lee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Charles B. Simone
- New York Proton Center, New York, NY 10035, USA
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Minglei Kang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA; (C.C.)
- New York Proton Center, New York, NY 10035, USA
| | - Hui Wu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450008, China
| |
Collapse
|
33
|
Guan F, Jiang D, Wang X, Yang M, Iga K, Li Y, Bronk L, Bronk J, Wang L, Guo Y, Sahoo N, Grosshans DR, Koong AC, Zhu XR, Mohan R. Mimicking large spot-scanning radiation fields for proton FLASH preclinical studies with a robotic motion platform. ARXIV 2024:arXiv:2409.09518v1. [PMID: 39314510 PMCID: PMC11419177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Previously, a synchrotron-based horizontal proton beamline (87.2 MeV) was successfully commissioned to deliver radiation doses in FLASH and conventional dose rate modes to small fields and volumes. In this study, we developed a strategy to increase the effective radiation field size using a custom robotic motion platform to automatically shift the positions of biological samples. The beam was first broadened with a thin tungsten scatterer and shaped by customized brass collimators for irradiating cell/organoid cultures in 96-well plates (a 7-mm-diameter circle) or for irradiating mice (1-cm2 square). Motion patterns of the robotic platform were written in G-code, with 9-mm spot spacing used for the 96-well plates and 10.6-mm spacing for the mice. The accuracy of target positioning was verified with a self-leveling laser system. The dose delivered in the experimental conditions was validated with EBT-XD film attached to the 96-well plate or the back of the mouse. Our film-measured dose profiles matched Monte Carlo calculations well (1D gamma pass rate >95%). The FLASH dose rates were 113.7 Gy/s for cell/organoid irradiation and 191.3 Gy/s for mouse irradiation. These promising results indicate that this robotic platform can be used to effectively increase the field size for preclinical experiments with proton FLASH.
Collapse
Affiliation(s)
- Fada Guan
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Dadi Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Xiaochun Wang
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Ming Yang
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Kiminori Iga
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
- Particle Therapy Division, Hitachi America, Ltd, 2535 Augustine Drive, Santa Clara, California 95054, USA
| | - Yuting Li
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Lawrence Bronk
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Julianna Bronk
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Liang Wang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Youming Guo
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Narayan Sahoo
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - David R. Grosshans
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Albert C. Koong
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Xiaorong R. Zhu
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Radhe Mohan
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| |
Collapse
|
34
|
Liu K, Titt U, Esplen N, Connell L, Konradsson E, Yang M, Wang X, Takaoka T, Li Z, Koong AC, Mitra D, Mohan R, Loo BW, Lin SH, Schüler E. Discordance in acute gastrointestinal toxicity between synchrotron-based proton and linac-based electron ultra-high dose rate irradiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.04.611307. [PMID: 39282305 PMCID: PMC11398481 DOI: 10.1101/2024.09.04.611307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
Purpose Proton FLASH has been investigated using cyclotron and synchrocyclotron beamlines but not synchrotron beamlines. We evaluated the impact of dose rate (ultra-high [UHDR] vs. conventional [CONV]) and beam configuration (shoot-through [ST] vs. spread-out-Bragg-peak [SOBP]) on acute radiation-induced gastrointestinal toxicity (RIGIT) in mice. We also compared RIGIT between synchrotron-based protons and linac-based electrons with matched mean dose rates. Methods and Materials We administered abdominal irradiation (12-14 Gy single fraction) to female C57BL/6J mice with an 87 MeV synchrotron-based proton beamline (2 cm diameter field size as a lateral beam). Dose rates were 0.2 Gy/s (S-T pCONV), 0.3 Gy/s (SOBP pCONV), 150 Gy/s (S-T pFLASH), and 230 Gy/s (SOBP pFLASH). RIGIT was assessed by the jejunal regenerating crypt assay and survival. We also compared responses to proton [pFLASH and pCONV] with responses to electron CONV (eCONV, 0.4 Gy/s) and electron FLASH (eFLASH, 188-205 Gy/s). Results The number of regenerating jejunal crypts at each matched dose was lowest for pFLASH (similar between S-T and SOBP), greater and similar between pCONV (S-T and SOBP) and eCONV, and greatest for eFLASH. Correspondingly, mice that received pFLASH SOBP had the lowest survival rates (50% at 50 days), followed by pFLASH S-T (80%), and pCONV SOBP (90%), but 100% of mice receiving pCONV S-T survived (log-rank P = 0.047 for the four groups). Conclusions Our findings are consistent with an increase in RIGIT after synchrotron-based pFLASH versus pCONV. This negative proton-specific FLASH effect versus linac-based electron irradiation underscores the importance of understanding the physical and biological factors that will allow safe and effective clinical translation.
Collapse
|
35
|
Ashraf MR, Melemenidis S, Liu K, Grilj V, Jansen J, Velasquez B, Connell L, Schulz JB, Bailat C, Libed A, Manjappa R, Dutt S, Soto L, Lau B, Garza A, Larsen W, Skinner L, Yu AS, Surucu M, Graves EE, Maxim PG, Kry SF, Vozenin MC, Schüler E, Loo BW. Multi-Institutional Audit of FLASH and Conventional Dosimetry With a 3D Printed Anatomically Realistic Mouse Phantom. Int J Radiat Oncol Biol Phys 2024; 120:287-300. [PMID: 38493902 DOI: 10.1016/j.ijrobp.2024.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 03/03/2024] [Accepted: 03/10/2024] [Indexed: 03/19/2024]
Abstract
PURPOSE We conducted a multi-institutional dosimetric audit between FLASH and conventional dose rate (CONV) electron irradiations by using an anatomically realistic 3-dimensional (3D) printed mouse phantom. METHODS AND MATERIALS A computed tomography (CT) scan of a live mouse was used to create a 3D model of bony anatomy, lungs, and soft tissue. A dual-nozzle 3D printer was used to print the mouse phantom using acrylonitrile butadiene styrene (∼1.02 g/cm3) and polylactic acid (∼1.24 g/cm3) simultaneously to simulate soft tissue and bone densities, respectively. The lungs were printed separately using lightweight polylactic acid (∼0.64 g/cm3). Hounsfield units (HU), densities, and print-to-print stability of the phantoms were assessed. Three institutions were each provided a phantom and each institution performed 2 replicates of irradiations at selected anatomic regions. The average dose difference between FLASH and CONV dose distributions and deviation from the prescribed dose were measured with radiochromic film. RESULTS Compared with the reference CT scan, CT scans of the phantom demonstrated mass density differences of 0.10 g/cm3 for bone, 0.12 g/cm3 for lung, and 0.03 g/cm3 for soft tissue regions. Differences in HU between phantoms were <10 HU for soft tissue and bone, with lung showing the most variation (54 HU), but with minimal effect on dose distribution (<0.5%). Mean differences between FLASH and CONV decreased from the first to the second replicate (4.3%-1.2%), and differences from the prescribed dose decreased for both CONV (3.6%-2.5%) and FLASH (6.4%-2.7%). Total dose accuracy suggests consistent pulse dose and pulse number, although these were not specifically assessed. Positioning variability was observed, likely due to the absence of robust positioning aids or image guidance. CONCLUSIONS This study marks the first dosimetric audit for FLASH using a nonhomogeneous phantom, challenging conventional calibration practices reliant on homogeneous phantoms. The comparison protocol offers a framework for credentialing multi-institutional studies in FLASH preclinical research to enhance reproducibility of biologic findings.
Collapse
Affiliation(s)
- M Ramish Ashraf
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Stavros Melemenidis
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Kevin Liu
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Veljko Grilj
- Institute of Radiation Physics, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Jeannette Jansen
- Radiation Oncology Laboratory, Department of Radiation Oncology, Lausanne, University Hospital and University of Lausanne, Switzerland
| | - Brett Velasquez
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Luke Connell
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joseph B Schulz
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Claude Bailat
- Institute of Radiation Physics, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Aaron Libed
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Rakesh Manjappa
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Suparna Dutt
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Luis Soto
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Brianna Lau
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Aaron Garza
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - William Larsen
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Lawrie Skinner
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Amy S Yu
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Murat Surucu
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Edward E Graves
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Peter G Maxim
- Department of Radiation Oncology, University of California, Irvine, California
| | - Stephen F Kry
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas; Imaging and Radiation Oncology Core, MD Anderson Cancer Center, Houston, USA
| | - Marie-Catherine Vozenin
- Radiation Oncology Laboratory, Department of Radiation Oncology, Lausanne, University Hospital and University of Lausanne, Switzerland; Radiotherapy and Radiobiology Sector, Radiation Therapy Service, University Hospital of Geneva, Geneva, Switzerland.
| | - Emil Schüler
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Billy W Loo
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
36
|
Hamatani N, Yagi M, Shimizu S, Ishino N, Shimizu M, Kuwana Y, Tsubouchi T, Takashina M, Miyoshi T, Nomura T, Toyoda T, Umezawa M, Nishio T, Koizumi M, Ogawa K, Kanai T. Investigation of Ionization Chamber Characteristics for Ultrahigh-dose-rate Scanned Carbon-ion Beams. In Vivo 2024; 38:2220-2227. [PMID: 39187321 PMCID: PMC11363756 DOI: 10.21873/invivo.13686] [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: 06/06/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND/AIM There are only a few studies on dosimetry with ultrahigh-dose-rate (uHDR) scanned carbon-ion beams. This study investigated the characteristics of four types of ionization chambers for the uHDR beam. MATERIALS AND METHODS We employed a newly developed large-plane parallel chamber to monitor a 208.3-MeV/u uHDR scanned carbon-ion beam with a 110-Gy/s average dose rate. The ionization chambers used were the Advanced Markus chamber (AMC), PinPoint 3D chamber (PPC), Farmer chamber (FC), and large-plane parallel chamber (StingRay). The AMC and StingRay surfaces and the PPC and FC geometric centers were aligned to the radiation isocenter using treatment room lasers. Using the voltage range stated in the instruction manuals, we obtained the saturation curves of the chambers. From these curves, we obtained the ion recombination correction factors using the two-voltage and three-voltage linear methods. The dose linearity was evaluated using five measurement points, and the chamber repeatability was verified by conducting repeated measurements for different dose values. RESULTS Although all chambers, except for AMC, reached saturation when specified voltages were applied, they exhibited excellent linearity for different dose values. The ion recombination correction factors of the AMC obtained using the aforementioned linear methods were nearly 1. Additionally, all chambers exhibited excellent repeatability. Although the standard deviation of the PPC for the lowest dose was ~1.5%, those of all the other chambers were <1.0%. CONCLUSION All ionization chambers can be used for measuring the relative dose, and absolute dose can be conveniently measured using the AMC with an uHDR carbon-ion scanned beam.
Collapse
Affiliation(s)
- Noriaki Hamatani
- Department of Medical Physics, Osaka Heavy-Ion Therapy Center, Osaka, Japan;
| | - Masashi Yagi
- Department of Carbon-Ion Radiotherapy, Graduate School of Medicine, Osaka University, Osaka, Japan;
| | - Shinichi Shimizu
- Department of Carbon-Ion Radiotherapy, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Naoki Ishino
- Medical Physics for Particle Therapy Laboratory, Department of Biophysical Engineering, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masaki Shimizu
- Healthcare Business Groupe, Hitachi High-Tech Corporation, Chiba, Japan
| | - Yoshiaki Kuwana
- Healthcare Business Groupe, Hitachi High-Tech Corporation, Chiba, Japan
| | - Toshiro Tsubouchi
- Department of Medical Physics, Osaka Heavy-Ion Therapy Center, Osaka, Japan
| | - Masaaki Takashina
- Department of Medical Physics, Osaka Heavy-Ion Therapy Center, Osaka, Japan
| | - Takuto Miyoshi
- Healthcare Business Groupe, Hitachi High-Tech Corporation, Chiba, Japan
| | - Takuya Nomura
- Healthcare Business Groupe, Hitachi High-Tech Corporation, Chiba, Japan
| | - Takashi Toyoda
- Healthcare Business Groupe, Hitachi High-Tech Corporation, Chiba, Japan
| | - Masumi Umezawa
- Healthcare Business Groupe, Hitachi High-Tech Corporation, Chiba, Japan
| | - Teiji Nishio
- Medical Physics Laboratory, Department of Biophysical Engineering, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masahiko Koizumi
- Radiation Oncology Laboratory, Department of Biophysical Engineering, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazuhiko Ogawa
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tatsuaki Kanai
- Medical Physics for Particle Therapy Laboratory, Department of Biophysical Engineering, Division of Health Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
37
|
Bookbinder A, Selvaraj B, Zhao X, Yang Y, Bell BI, Pennock M, Tsai P, Tomé WA, Isabelle Choi J, Lin H, Simone CB, Guha C, Kang M. Validation and reproducibility of in vivo dosimetry for pencil beam scanned FLASH proton treatment in mice. Radiother Oncol 2024; 198:110404. [PMID: 38942121 DOI: 10.1016/j.radonc.2024.110404] [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: 03/14/2024] [Revised: 06/07/2024] [Accepted: 06/19/2024] [Indexed: 06/30/2024]
Abstract
PURPOSE To investigate quality assurance (QA) techniques for in vivo dosimetry and establish its routine uses for proton FLASH small animal experiments with a saturated monitor chamber. METHODS AND MATERIALS 227 mice were irradiated at FLASH or conventional (CONV) dose rates with a 250 MeV FLASH-capable proton beamline using pencil beam scanning to characterize the proton FLASH effect on abdominal irradiation and examining various endpoints. A 2D strip ionization chamber array (SICA) detector was positioned upstream of collimation and used for in vivo dose monitoring during irradiation. Before each irradiation series, SICA signal was correlated with the isocenter dose at each delivered dose rate. Dose, dose rate, and 2D dose distribution for each mouse were monitored with the SICA detector. RESULTS Calibration curves between the upstream SICA detector signal and the delivered dose at isocenter had good linearity with minimal R2 values of 0.991 (FLASH) and 0.985 (CONV), and slopes were consistent for each modality. After reassigning mice, standard deviations were less than 1.85 % (FLASH) and 0.83 % (CONV) for all dose levels, with no individual subject dose falling outside a ± 3.6 % range of the designated dose. FLASH fields had a field-averaged dose rate of 79.0 ± 0.8 Gy/s and mean local average dose rate of 160.6 ± 3.0 Gy/s. In vivo dosimetry allowed for the accurate detection of variation between the delivered and the planned dose. CONCLUSION In vivo dosimetry benefits FLASH experiments through enabling real-time dose and dose rate monitoring allowing mouse cohort regrouping when beam fluctuation causes delivered dose to vary from planned dose.
Collapse
Affiliation(s)
| | | | | | - Yunjie Yang
- New York Proton Center, New York, NY, USA; Departments of Radiation Oncology and Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brett I Bell
- Department of Radiation Oncology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA; Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Michael Pennock
- Department of Radiation Oncology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA
| | - Pingfang Tsai
- New York Proton Center, New York, NY, USA; Departments of Radiation Oncology and Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wolfgang A Tomé
- Department of Radiation Oncology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA; Institute for Onco-Physics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - J Isabelle Choi
- New York Proton Center, New York, NY, USA; Departments of Radiation Oncology and Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Haibo Lin
- New York Proton Center, New York, NY, USA; Departments of Radiation Oncology and Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiation Oncology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA
| | - Charles B Simone
- New York Proton Center, New York, NY, USA; Departments of Radiation Oncology and Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chandan Guha
- Department of Radiation Oncology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA; Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA; Institute for Onco-Physics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Minglei Kang
- New York Proton Center, New York, NY, USA; Departments of Radiation Oncology and Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
38
|
Dai T, Sloop AM, Ashraf MR, Sunnerberg JP, Clark MA, Bruza P, Pogue BW, Jarvis L, Gladstone DJ, Zhang R. Commissioning an ultra-high-dose-rate electron linac with end-to-end tests. Phys Med Biol 2024; 69:165028. [PMID: 39084661 DOI: 10.1088/1361-6560/ad69fc] [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: 02/28/2024] [Accepted: 07/31/2024] [Indexed: 08/02/2024]
Abstract
Objective. The FLASH effect can potentially be used to improve the therapeutic ratio of radiotherapy (RT) through delivery of Ultra-high-dose-rate (UHDR) irradiation. Research is actively being conducted to translate UHDR-RT and for this purpose the Mobetron is capable of producing electron beams at both UHDR and conventional dose rates for FLASH research and translation. This work presents commissioning of an UHDR Mobetron with end-to-end tests developed for preclinical research.Approach. UHDR electron beams were commissioned with an efficient approach utilizing a 3D-printed water tank and film to fully characterize beam characteristics and dependences on field size, pulse width (PW) and pulse repetition frequency (PRF). This commissioning data was used to implement a beam model using the GAMOS Monte Carlo toolkit for the preclinical research. Then, the workflow for preclinical FLASH irradiation was validated with end-to-end tests delivered to a 3D-printed mouse phantom with internal inhomogeneities.Main results.PDDs, profiles and output factors acquired with radiochromic films were precisely measured, with a PRF that showed little effect on the UHDR beam energy and spatial characteristics. Increasing PW reduced theDmaxand R50by 2.08 mmµs-1and 1.28 mmµs-1respectively. An end-to-end test of the preclinical research workflow showed that both profiles in head-foot and lateral directions were in good agreement with the MC calculations for the heterogeneous 3D printed mouse phantom with Gamma index above 93% for 2 mm/2% criteria, and 99% for 3 mm/3%.Significance. The UHDR Mobetron is a versatile tool for FLASH preclinical research and this comprehensive beam model and workflow was validated to meet the requirements for conducting translational FLASH research.
Collapse
Affiliation(s)
- Tianyuan Dai
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States of America
- Department of Radiation Oncology Physics and Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250000, People's Republic of China
| | - Austin M Sloop
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States of America
| | - Muhammad R Ashraf
- Stanford Radiation Oncology, Palo Alto, CA 94304, United States of America
| | - Jacob P Sunnerberg
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States of America
| | - Megan A Clark
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States of America
| | - Petr Bruza
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States of America
| | - Brian W Pogue
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States of America
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756, United States of America
- Department of Medical Physics, Wisconsin Institutes for Medical Research, University of Wisconsin, Madison, WI 53705, United States of America
| | - Lesley Jarvis
- Department of Medicine, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, United States of America
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756, United States of America
| | - David J Gladstone
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States of America
- Department of Medicine, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, United States of America
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756, United States of America
| | - Rongxiao Zhang
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, United States of America
- Department of Radiation Medicine, New York Medical College, Valhalla, NY 10595, United States of America
| |
Collapse
|
39
|
Tsai P, Yang Y, Wu M, Chen C, Yu F, Simone CB, Choi JI, Tomé WA, Lin H. A comprehensive pre-clinical treatment quality assurance program using unique spot patterns for proton pencil beam scanning FLASH radiotherapy. J Appl Clin Med Phys 2024; 25:e14400. [PMID: 38831639 PMCID: PMC11302823 DOI: 10.1002/acm2.14400] [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: 01/03/2024] [Revised: 03/14/2024] [Accepted: 05/07/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Quality assurance (QA) for ultra-high dose rate (UHDR) irradiation is a crucial aspect in the emerging field of FLASH radiotherapy (FLASH-RT). This innovative treatment approach delivers radiation at UHDR, demanding careful adoption of QA protocols and procedures. A comprehensive understanding of beam properties and dosimetry consistency is vital to ensure the safe and effective delivery of FLASH-RT. PURPOSE To develop a comprehensive pre-treatment QA program for cyclotron-based proton pencil beam scanning (PBS) FLASH-RT. Establish appropriate tolerances for QA items based on this study's outcomes and TG-224 recommendations. METHODS A 250 MeV proton spot pattern was designed and implemented using UHDR with a 215nA nozzle beam current. The QA pattern that covers a central uniform field area, various spot spacings, spot delivery modes and scanning directions, and enabling the assessment of absolute, relative and temporal dosimetry QA parameters. A strip ionization chamber array (SICA) and an Advanced Markus chamber were utilized in conjunction with a 2 cm polyethylene slab and a range (R80) verification wedge. The data have been monitored for over 3 months. RESULTS The relative dosimetries were compliant with TG-224. The variations of temporal dosimetry for scanning speed, spot dwell time, and spot transition time were within ± 1 mm/ms, ± 0.2 ms, and ± 0.2 ms, respectively. While the beam-to-beam absolute output on the same day reached up to 2.14%, the day-to-day variation was as high as 9.69%. High correlation between the absolute dose and dose rate fluctuations were identified. The dose rate of the central 5 × 5 cm2 field exhibited variations within 5% of the baseline value (155 Gy/s) during an experimental session. CONCLUSIONS A comprehensive QA program for FLASH-RT was developed and effectively assesses the performance of a UHDR delivery system. Establishing tolerances to unify standards and offering direction for future advancements in the evolving FLASH-RT field.
Collapse
Affiliation(s)
| | - Yunjie Yang
- Department of Radiation OncologyMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Mengjou Wu
- New York Proton CenterNew YorkNew YorkUSA
| | | | - Francis Yu
- New York Proton CenterNew YorkNew YorkUSA
| | | | | | - Wolfgang A. Tomé
- Department of Radiation OncologyMontefiore Medical Center and Albert Einstein College of MedicineBronxNew YorkUSA
| | - Haibo Lin
- New York Proton CenterNew YorkNew YorkUSA
- Department of Radiation OncologyMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
- Department of Radiation OncologyMontefiore Medical Center and Albert Einstein College of MedicineBronxNew YorkUSA
| |
Collapse
|
40
|
Held KD, McNamara AL, Daartz J, Bhagwat MS, Rothwell B, Schuemann J. Dose Rate Effects from the 1950s through to the Era of FLASH. Radiat Res 2024; 202:161-176. [PMID: 38954556 PMCID: PMC11426361 DOI: 10.1667/rade-24-00024.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/09/2024] [Indexed: 07/04/2024]
Abstract
Numerous dose rate effects have been described over the past 6-7 decades in the radiation biology and radiation oncology literature depending on the dose rate range being discussed. This review focuses on the impact and understanding of altering dose rates in the context of radiation therapy, but does not discuss dose rate effects as relevant to radiation protection. The review starts with a short historic review of early studies on dose rate effects, considers mechanisms thought to underlie dose rate dependencies, then discusses some current issues in clinical findings with altered dose rates, the importance of dose rate in brachytherapy, and the current timely topic of the use of very high dose rates, so-called FLASH radiotherapy. The discussion includes dose rate effects in vitro in cultured cells, in in vivo experimental systems and in the clinic, including both tumors and normal tissues. Gaps in understanding dose rate effects are identified, as are opportunities for improving clinical use of dose rate modulation.
Collapse
Affiliation(s)
- Kathryn D Held
- Department of Radiation Oncology, Massachusetts General Hospital Hospital/Harvard Medical School, Boston, Massachusetts 02114
- National Council on Radiation Protection and Measurements, Bethesda, Maryland 20814
| | - Aimee L McNamara
- Department of Radiation Oncology, Massachusetts General Hospital Hospital/Harvard Medical School, Boston, Massachusetts 02114
| | - Juliane Daartz
- Department of Radiation Oncology, Massachusetts General Hospital Hospital/Harvard Medical School, Boston, Massachusetts 02114
| | - Mandar S Bhagwat
- Department of Radiation Oncology, Massachusetts General Hospital Hospital/Harvard Medical School, Boston, Massachusetts 02114
| | - Bethany Rothwell
- Department of Radiation Oncology, Massachusetts General Hospital Hospital/Harvard Medical School, Boston, Massachusetts 02114
| | - Jan Schuemann
- Department of Radiation Oncology, Massachusetts General Hospital Hospital/Harvard Medical School, Boston, Massachusetts 02114
| |
Collapse
|
41
|
Ma J, Lin Y, Tang M, Zhu YN, Gan GN, Rotondo RL, Chen RC, Gao H. Simultaneous dose and dose rate optimization via dose modifying factor modeling for FLASH effective dose. Med Phys 2024; 51:5190-5203. [PMID: 38873848 PMCID: PMC11783338 DOI: 10.1002/mp.17251] [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: 02/15/2024] [Revised: 04/28/2024] [Accepted: 05/31/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Although the FLASH radiotherapy (FLASH) can improve the sparing of organs-at-risk (OAR) via the FLASH effect, it is generally a tradeoff between the physical dose coverage and the biological FLASH coverage, for which the concept of FLASH effective dose (FED) is needed to quantify the net improvement of FLASH, compared to the conventional radiotherapy (CONV). PURPOSE This work will develop the first-of-its-kind treatment planning method called simultaneous dose and dose rate optimization via dose modifying factor modeling (SDDRO-DMF) for proton FLASH that directly optimizes FED. METHODS SDDRO-DMF models and optimizes FED using FLASH dose modifying factor (DMF) models, which can be classified into two categories: (1) the phenomenological model of the FLASH effect, such as the FLASH effectiveness model (FEM); (2) the mechanistic model of the FLASH radiobiology, such as the radiolytic oxygen depletion (ROD) model. The general framework of SDDRO-DMF will be developed, with specific DMF models using FEM and ROD, as a demonstration of general applicability of SDDRO-DMF for proton FLASH via transmission beams (TB) or Bragg peaks (BP) with single-field or multi-field irradiation. The FLASH dose rate is modeled as pencil beam scanning dose rate. The solution algorithm for solving the inverse optimization problem of SDDRO-DMF is based on iterative convex relaxation method. RESULTS SDDRO-DMF is validated in comparison with IMPT and a state-of-the-art method called SDDRO, with demonstrated efficacy and improvement for reducing the high dose and the high-dose volume for OAR in terms of FED. For example, in a SBRT lung case of the dose-limiting factor that the max dose of brachial plexus should be no more than 26 Gy, only SDDRO-DMF met this max dose constraint; moreover, SDDRO-DMF completely eliminated the high-dose (V70%) volume to zero for CTV10mm (a high-dose region as a 10 mm ring expansion of CTV). CONCLUSION We have proposed a new proton FLASH optimization method called SDDRO-DMF that directly optimizes FED using phenomenological or mechanistic models of DMF, and have demonstrated the efficacy of SDDO-DMF in reducing the high-dose volume or/and the high-dose value for OAR, compared to IMPT and a state-of-the-art method SDDRO.
Collapse
Affiliation(s)
- Jiangjun Ma
- Institute of Natural Sciences and School of Mathematics, Shanghai Jiao Tong University, Shanghai, China
| | - Yuting Lin
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas city, Kansas, USA
| | - Min Tang
- Institute of Natural Sciences and School of Mathematics, Shanghai Jiao Tong University, Shanghai, China
| | - Ya-Nan Zhu
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas city, Kansas, USA
| | - Gregory N Gan
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas city, Kansas, USA
| | - Ronny L Rotondo
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas city, Kansas, USA
| | - Ronald C Chen
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas city, Kansas, USA
| | - Hao Gao
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas city, Kansas, USA
| |
Collapse
|
42
|
Portier L, Daira P, Fourmaux B, Heinrich S, Becerra M, Fouillade C, Berthault N, Dutreix M, Londoño-Vallejo A, Verrelle P, Bernoud-Hubac N, Favaudon V. Differential Remodeling of the Oxylipin Pool After FLASH Versus Conventional Dose-Rate Irradiation In Vitro and In Vivo. Int J Radiat Oncol Biol Phys 2024; 119:1481-1492. [PMID: 38340776 DOI: 10.1016/j.ijrobp.2024.01.210] [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: 10/06/2023] [Revised: 01/09/2024] [Accepted: 01/13/2024] [Indexed: 02/12/2024]
Abstract
PURPOSE The products of lipid peroxidation have been implicated in human diseases and aging. This prompted us to investigate the response to conventional (CONV) versus FLASH irradiation of oxylipins, a family of bioactive lipid metabolites derived from omega-3 or omega-6 polyunsaturated fatty acids through oxygen-dependent non-enzymatic as well as dioxygenase-mediated free radical reactions. METHODS AND MATERIALS Ultrahigh performance liquid chromatography coupled to tandem mass spectrometry was used to quantify the expression of 37 oxylipins derived from eicosatetraenoic, eicosapentaenoic and docosahexaenoic acid in mouse lung and in normal or cancer cells exposed to either radiation modality under precise monitoring of the temperature and oxygenation. Among the 37 isomers assayed, 14-16 were present in high enough amount to enable quantitative analysis. The endpoints were the expression of oxylipins as a function of the dose of radiation, normoxia versus hypoxia, temperature and post-irradiation time. RESULTS In normal, normoxic cells at 37°C radiation elicited destruction and neosynthesis of oxylipins acting antagonistically on a background subject to rapid remodeling by oxygenases. Neosynthesis was observed in the CONV mode only, in such a way that the level of oxylipins at 5 minutes after FLASH irradiation was 20-50% lower than in non-irradiated and CONV-irradiated cells. Hypoxia mitigated the differential CONV versus FLASH response in some oxylipins. These patterns were not reproduced in tumor cells. Depression of specific oxylipins following FLASH irradiation was observed in mouse lung at 5 min following irradiation, with near complete recovery in 24 hours and further remodeling at one week and two months post-irradiation. CONCLUSIONS Down-regulation of oxylipins was a hallmark of FLASH irradiation specific of normal cells. Temperature effects suggest that this process occurs via diffusion-controlled, bimolecular recombination of a primary radical species upstream from peroxyl radical formation and evoke a major role of the membrane composition and fluidity in response to the FLASH modality.
Collapse
Affiliation(s)
- Lucie Portier
- Institut Curie, Research Division, Inserm U 1021-CNRS UMR 3347, Paris-Saclay University, PSL Research University, Centre Universitaire CS 90030, Orsay, France
| | - Patricia Daira
- Univ Lyon, INSA Lyon, CNRS, LaMCoS, UMR 5259, Villeurbanne, France
| | | | - Sophie Heinrich
- Institut Curie, Research Division, Inserm U 1021-CNRS UMR 3347, Paris-Saclay University, PSL Research University, Centre Universitaire CS 90030, Orsay, France
| | - Margaux Becerra
- Institut Curie, Research Division, Inserm U 1021-CNRS UMR 3347, Paris-Saclay University, PSL Research University, Centre Universitaire CS 90030, Orsay, France
| | - Charles Fouillade
- Institut Curie, Research Division, Inserm U 1021-CNRS UMR 3347, Paris-Saclay University, PSL Research University, Centre Universitaire CS 90030, Orsay, France
| | - Nathalie Berthault
- Institut Curie, Research Division, Inserm U 1021-CNRS UMR 3347, Paris-Saclay University, PSL Research University, Centre Universitaire CS 90030, Orsay, France
| | - Marie Dutreix
- Institut Curie, Research Division, Inserm U 1021-CNRS UMR 3347, Paris-Saclay University, PSL Research University, Centre Universitaire CS 90030, Orsay, France
| | - Arturo Londoño-Vallejo
- Institut Curie, Research Division, Inserm U 1021-CNRS UMR 3347, Paris-Saclay University, PSL Research University, Centre Universitaire CS 90030, Orsay, France
| | - Pierre Verrelle
- Institut Curie, Hospital Section, Department of Radiotherapy-Oncology, 26 rue d'Ulm, 75248 Paris Cedex 05, France; Institut Curie, Research Division, Inserm U 1196-CNRS UMR 9187, Paris-Saclay University, PSL Research University, Centre Universitaire CS 90030, Orsay, France
| | | | - Vincent Favaudon
- Institut Curie, Research Division, Inserm U 1021-CNRS UMR 3347, Paris-Saclay University, PSL Research University, Centre Universitaire CS 90030, Orsay, France.
| |
Collapse
|
43
|
Amit U, Uslu U, Verginadis II, Kim MM, Motlagh SAO, Diffenderfer ES, Assenmacher CA, Bicher S, Atoche SJ, Ben-Josef E, Young RM, June CH, Koumenis C. Proton radiation boosts the efficacy of mesothelin-targeting chimeric antigen receptor T cell therapy in pancreatic cancer. Proc Natl Acad Sci U S A 2024; 121:e2403002121. [PMID: 39047033 PMCID: PMC11294999 DOI: 10.1073/pnas.2403002121] [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: 02/12/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents a challenge in oncology, with limited treatment options for advanced-stage patients. Chimeric antigen receptor T cell (CAR T) therapy targeting mesothelin (MSLN) shows promise, but challenges such as the hostile immunosuppressive tumor microenvironment (TME) hinder its efficacy. This study explores the synergistic potential of combining proton radiation therapy (RT) with MSLN-targeting CAR T therapy in a syngeneic PDAC model. Proton RT significantly increased MSLN expression in tumor cells and caused a significant increase in CAR T cell infiltration into tumors. The combination therapy reshaped the immunosuppressive TME, promoting antitumorigenic M1 polarized macrophages and reducing myeloid-derived suppressor cells (MDSC). In a flank PDAC model, the combination therapy demonstrated superior attenuation of tumor growth and improved survival compared to individual treatments alone. In an orthotopic PDAC model treated with image-guided proton RT, tumor growth was significantly reduced in the combination group compared to the RT treatment alone. Further, the combination therapy induced an abscopal effect in a dual-flank tumor model, with increased serum interferon-γ levels and enhanced proliferation of extratumoral CAR T cells. In conclusion, combining proton RT with MSLN-targeting CAR T therapy proves effective in modulating the TME, enhancing CAR T cell trafficking, and exerting systemic antitumor effects. Thus, this combinatorial approach could present a promising strategy for improving outcomes in unresectable PDAC.
Collapse
Affiliation(s)
- Uri Amit
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Department of Radiation Oncology, Tel Aviv Medical Center, Tel Aviv64239, Israel
| | - Ugur Uslu
- Department of Pathology and Laboratory Medicine, Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA19104
| | - Ioannis I. Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Michele M. Kim
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Seyyedeh Azar Oliaei Motlagh
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Eric S. Diffenderfer
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Charles-Antoine Assenmacher
- Department of Pathobiology, School of Veterinary Medicine, Comparative Pathology Core, University of Pennsylvania, Philadelphia, PA19104
| | - Sandra Bicher
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Sebastian J. Atoche
- Department of Pathology and Laboratory Medicine, Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA19104
| | - Edgar Ben-Josef
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Regina M. Young
- Department of Pathology and Laboratory Medicine, Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA19104
| | - Carl H. June
- Department of Pathology and Laboratory Medicine, Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA19104
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| |
Collapse
|
44
|
Kim K, Kim MM, Skoufos G, Diffenderfer ES, Motlagh SAO, Kokkorakis M, Koliaki I, Morcos G, Shoniyozov K, Griffin J, Hatzigeorgiou AG, Metz JM, Lin A, Feigenberg SJ, Cengel KA, Ky B, Koumenis C, Verginadis II. 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 2024; 119:1234-1247. [PMID: 38364948 PMCID: PMC11209795 DOI: 10.1016/j.ijrobp.2024.01.224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 01/12/2024] [Accepted: 01/28/2024] [Indexed: 02/18/2024]
Abstract
PURPOSE Studies during the past 9 years suggest that delivering radiation at dose rates exceeding 40 Gy/s, known as "FLASH" radiation therapy, enhances the therapeutic index of radiation therapy (RT) by decreasing normal tissue damage while maintaining tumor response compared with conventional (or standard) RT. This study demonstrates the cardioprotective benefits of FLASH proton RT (F-PRT) compared with standard (conventional) proton RT (S-PRT), as evidenced by reduced acute and chronic cardiac toxicities. METHODS AND MATERIALS Mice were imaged using cone beam computed tomography to precisely determine the heart's apex as the beam isocenter. Irradiation was conducted using a shoot-through technique with a 5-mm diameter circular collimator. Bulk RNA-sequencing was performed on nonirradiated samples, as well as apexes treated with F-PRT or S-PRT, at 2 weeks after a single 40 Gy dose. Inflammatory responses were assessed through multiplex cytokine/chemokine microbead assay and immunofluorescence analyses. Levels of perivascular fibrosis were quantified using Masson's Trichrome and Picrosirius red staining. Additionally, cardiac tissue functionality was evaluated by 2-dimensional echocardiograms at 8- and 30-weeks post-PRT. RESULTS Radiation damage was specifically localized to the heart's apex. RNA profiling of cardiac tissues treated with PRT revealed that S-PRT uniquely upregulated pathways associated with DNA damage response, induction of tumor necrosis factor superfamily, and inflammatory response, and F-PRT primarily affected cytoplasmic translation, mitochondrion organization, and adenosine triphosphate synthesis. Notably, F-PRT led to a milder inflammatory response, accompanied by significantly attenuated changes in transforming growth factor β1 and α smooth muscle actin levels. Critically, F-PRT decreased collagen deposition and better preserved cardiac functionality compared with S-PRT. CONCLUSIONS This study demonstrated that F-PRT reduces the induction of an inflammatory environment with lower expression of inflammatory cytokines and profibrotic factors. Importantly, the results indicate that F-PRT better preserves cardiac functionality, as confirmed by echocardiography analysis, while also mitigating the development of long-term fibrosis.
Collapse
Affiliation(s)
- Kyle Kim
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michele M Kim
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Giorgos Skoufos
- Department of Electrical & Computer Engineering, University of Thessaly, Greece; Hellenic Pasteur Institute, Athens, Greece
| | - Eric S Diffenderfer
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Seyyedeh Azar Oliaei Motlagh
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michail Kokkorakis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ilektra Koliaki
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - George Morcos
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Khayrullo Shoniyozov
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joanna Griffin
- Department of Medicine, Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Artemis G Hatzigeorgiou
- Department of Electrical & Computer Engineering, University of Thessaly, Greece; Hellenic Pasteur Institute, Athens, Greece; DIANA-Laboratory, Department of Computer Science and Biomedical Informatics, University of Thessaly, Thessaly, Greece
| | - James M Metz
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alexander Lin
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Steven J Feigenberg
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Keith A Cengel
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bonnie Ky
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Ioannis I Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
45
|
Verginadis II, Velalopoulou A, Kim MM, Kim K, Paraskevaidis I, Bell B, Oliaei Motlagh SA, Karaj A, Banerjee E, Finesso G, Assenmacher CA, Radaelli E, Lu J, Lin Y, Putt ME, Diffenderfer ES, Guha C, Qin L, Metz JM, Maity A, Cengel KA, Koumenis C, Busch TM. FLASH proton reirradiation, with or without hypofractionation, mitigates chronic toxicity in the normal murine intestine, skin, and bone. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602528. [PMID: 39026805 PMCID: PMC11257476 DOI: 10.1101/2024.07.08.602528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Background and purpose The normal tissue sparing afforded by FLASH radiotherapy (RT) is being intensely investigated for potential clinical translation. Here, we studied the effects of FLASH proton RT (F-PRT) in the reirradiation setting, with or without hypofractionation. Chronic toxicities in three murine models of normal tissue toxicity including the intestine, skin, and bone were investigated. Materials and methods In studies of the intestine, single-dose irradiation was performed with 12 Gy of Standard proton RT (S-PRT), followed by a second dose of 12 Gy of F-PRT or S-PRT. Additionally, a hypofractionation scheme was applied in the reirradiation setting (3 x 6.4 Gy of F-PRT or S-PRT, given every 48 hrs). In studies of skin/bone of the murine leg, 15 Gy of S-PRT was followed by hypofractionated reirradiation with F-PRT or S-PRT (3 x 11 Gy). Results Compared to reirradiation with S-PRT, F-PRT reduced intestinal fibrosis and collagen deposition in the reirradiation setting and significantly increased survival rate, demonstrating its protective effects on intestinal tissues. In previously irradiated leg tissues, reirradiation with hypofractionated F-PRT created transient dermatitis that fully resolved in contrast to reirradiation with hypofractionated S-PRT. Lymphedema was also alleviated after a second course of radiation with F-PRT, along with significant reductions in the accumulation of fibrous connective tissue in the skin compared to mice reirradiated with S-PRT. The delivery of a second course of fractionated S-PRT induced tibial fractures in 83.3% of the mice, whereas only 20% of mice reirradiated with F-PRT presented with fractures. Conclusion These studies provide the first evidence of the sparing effects of F-PRT, in the setting of hypofractionated reirradiation. The results support FLASH as highly relevant to the reirradiation regimen where it exhibits significant potential to minimize chronic complications for patients undergoing RT.
Collapse
|
46
|
Chow JCL, Ruda HE. Impact of Scattering Foil Composition on Electron Energy Distribution in a Clinical Linear Accelerator Modified for FLASH Radiotherapy: A Monte Carlo Study. MATERIALS (BASEL, SWITZERLAND) 2024; 17:3355. [PMID: 38998435 PMCID: PMC11243336 DOI: 10.3390/ma17133355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/14/2024]
Abstract
This study investigates how scattering foil materials and sampling holder placement affect electron energy distribution in electron beams from a modified medical linear accelerator for FLASH radiotherapy. We analyze electron energy spectra at various positions-ionization chamber, mirror, and jaw-to evaluate the impact of Cu, Pb-Cu, Pb, and Ta foils. Our findings show that close proximity to the source intensifies the dependence of electron energy distribution on foil material, enabling precise beam control through material selection. Monte Carlo simulations are effective for designing foils to achieve desired energy distributions. Moving the sampling holder farther from the source reduces foil material influence, promoting more uniform energy spreads, particularly in the 0.5-10 MeV range for 12 MeV electron beams. These insights emphasize the critical role of tailored material selection and sampling holder positioning in optimizing electron energy distribution and fluence intensity for FLASH radiotherapy research, benefiting both experimental design and clinical applications.
Collapse
Affiliation(s)
- James C L Chow
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1X6, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - Harry E Ruda
- Centre of Advance Nanotechnology, Faculty of Applied Science and Engineering, University of Toronto, Toronto, ON M5S 3E4, Canada
- Department of Materials Science and Engineering, University of Toronto, Toronto, ON M5S 3E4, Canada
| |
Collapse
|
47
|
Kristensen L, Poulsen PR, Kanouta E, Rohrer S, Ankjærgaard C, Andersen CE, Johansen JG, Simeonov Y, Weber U, Grau C, Sørensen BS. Spread-out Bragg peak FLASH: quantifying normal tissue toxicity in a murine model. Front Oncol 2024; 14:1427667. [PMID: 39026976 PMCID: PMC11256197 DOI: 10.3389/fonc.2024.1427667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 06/18/2024] [Indexed: 07/20/2024] Open
Abstract
Objective A favorable effect of ultra-high dose rate (FLASH) radiation on normal tissue-sparing has been indicated in several preclinical studies. In these studies, the adverse effects of radiation damage were reduced without compromising tumor control. Most studies of proton FLASH investigate these effects within the entrance of a proton beam. However, the real advantage of proton therapy lies in the Spread-out Bragg Peak (SOBP), which allows for giving a high dose to a target with a limited dose to healthy tissue at the entrance of the beam. Therefore, a clinically relevant investigation of the FLASH effect would be of healthy tissues within a SOBP. Our study quantified the tissue-sparing effect of FLASH radiation on acute and late toxicity within an SOBP in a murine model. Material/Methods Radiation-induced damage was assessed for acute and late toxicity in the same mice following irradiation with FLASH (Field dose rate of 60 Gy/s) or conventional (CONV, 0.34 Gy/s) dose rates. The right hindleg of unanesthetized female CDF1 mice was irradiated with single-fraction doses between 19.9-49.7 Gy for CONV and 30.4-65.9 Gy for FLASH with 5-8 mice per dose. The leg was placed in the middle of a 5 cm SOBP generated from a mono-energetic beam using a 2D range modulator. Acute skin toxicity quantified by hair loss, moist desquamation and toe separation was monitored daily within 29 days post-treatment. Late toxicity of fibrotic development measured by leg extendibility was monitored biweekly until 30 weeks post-treatment. Results Comparison of acute skin toxicity following radiation indicated a tissue-sparing effect of FLASH compared to conventional single-fraction radiation with a mean protection ratio of 1.40 (1.35-1.46). Fibrotic development similarly indicated normal tissue sparing with a 1.18 (1.17-1.18) protection ratio. The acute skin toxicity tissue sparing was similar to data from entrance-beam irradiations of Sørensen et al. (4). Conclusion Full dose-response curves for acute and late toxicity after CONV and FLASH radiation were obtained. Radiation within the SOBP retains the normal-tissue-sparing effect of FLASH with a dose-modifying factor of 40% for acute skin damage and 18% for fibrotic development.
Collapse
Affiliation(s)
- Line Kristensen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Per Rugaard Poulsen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Eleni Kanouta
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Sky Rohrer
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
| | | | | | - Jacob G. Johansen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Yuri Simeonov
- Institut für Medizinische Physik und Strahlenschutz, Technische Hochschule Mittelhessen, Giessen, Germany
| | - Uli Weber
- Department for Biophysics, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Cai Grau
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Brita Singers Sørensen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
48
|
Fenwick JD, Mayhew C, Jolly S, Amos RA, Hawkins MA. Navigating the straits: realizing the potential of proton FLASH through physics advances and further pre-clinical characterization. Front Oncol 2024; 14:1420337. [PMID: 39022584 PMCID: PMC11252699 DOI: 10.3389/fonc.2024.1420337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/11/2024] [Indexed: 07/20/2024] Open
Abstract
Ultra-high dose-rate 'FLASH' radiotherapy may be a pivotal step forward for cancer treatment, widening the therapeutic window between radiation tumour killing and damage to neighbouring normal tissues. The extent of normal tissue sparing reported in pre-clinical FLASH studies typically corresponds to an increase in isotoxic dose-levels of 5-20%, though gains are larger at higher doses. Conditions currently thought necessary for FLASH normal tissue sparing are a dose-rate ≥40 Gy s-1, dose-per-fraction ≥5-10 Gy and irradiation duration ≤0.2-0.5 s. Cyclotron proton accelerators are the first clinical systems to be adapted to irradiate deep-seated tumours at FLASH dose-rates, but even using these machines it is challenging to meet the FLASH conditions. In this review we describe the challenges for delivering FLASH proton beam therapy, the compromises that ensue if these challenges are not addressed, and resulting dosimetric losses. Some of these losses are on the same scale as the gains from FLASH found pre-clinically. We therefore conclude that for FLASH to succeed clinically the challenges must be systematically overcome rather than accommodated, and we survey physical and pre-clinical routes for achieving this.
Collapse
Affiliation(s)
- John D. Fenwick
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Christopher Mayhew
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Simon Jolly
- Department of Physics and Astronomy, University College London, London, United Kingdom
| | - Richard A. Amos
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Maria A. Hawkins
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
- Clinical Oncology, Radiotherapy Department, University College London NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
49
|
Mossahebi S, Byrne K, Jiang K, Gerry A, Deng W, Repetto C, Jackson IL, Sawant A, Poirier Y. A high-throughput focused collimator for OAR-sparing preclinical proton FLASH studies: commissioning and validation. Phys Med Biol 2024; 69:14NT01. [PMID: 38876112 DOI: 10.1088/1361-6560/ad589f] [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: 03/23/2024] [Accepted: 06/14/2024] [Indexed: 06/16/2024]
Abstract
Objective. To fabricate and validate a novel focused collimator designed to spare normal tissue in a murine hemithoracic irradiation model using 250 MeV protons delivered at ultra-high dose rates (UHDRs) for preclinical FLASH radiation therapy (FLASH-RT) studies.Approach. A brass collimator was developed to shape 250 MeV UHDR protons from our Varian ProBeam. Six 13 mm apertures, of equivalent size to kV x-ray fields historically used to perform hemithorax irradiations, were precisely machined to match beam divergence, allowing concurrent hemithoracic irradiation of six mice while sparing the contralateral lung and abdominal organs. The collimated field profiles were characterized by film dosimetry, and a radiation survey of neutron activation was performed to ensure the safety of staff positioning animals.Main results. The brass collimator produced 1.2 mm penumbrae radiation fields comparable to kV x-rays used in preclinical studies. The penumbrae in the six apertures are similar, with full-width half-maxima of 13.3 mm and 13.5 mm for the central and peripheral apertures, respectively. The collimator delivered a similar dose at an average rate of 52 Gy s-1for all apertures. While neutron activation produces a high (0.2 mSv h-1) initial ambient equivalent dose rate, a parallel work-flow in which imaging and setup are performed without the collimator ensures safety to staff.Significance. Scanned protons have the greatest potential for future translation of FLASH-RT in clinical treatments due to their ability to treat deep-seated tumors with high conformality. However, the Gaussian distribution of dose in proton spots produces wider lateral penumbrae compared to other modalities. This presents a challenge in small animal pre-clinical studies, where millimeter-scale penumbrae are required to precisely target the intended volume. Offering high-throughput irradiation of mice with sharp penumbrae, our novel collimator-based platform serves as an important benchmark for enabling large-scale, cost-effective radiobiological studies of the FLASH effect in murine models.
Collapse
Affiliation(s)
- Sina Mossahebi
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Kevin Byrne
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Kai Jiang
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Andrew Gerry
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Wei Deng
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Carlo Repetto
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Isabel L Jackson
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, United States of America
- True North Biopharm, LLC, Rockville, MD, United States of America
| | - Amit Sawant
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Yannick Poirier
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
50
|
Fu J, Yang Z, Melemenidis S, Viswanathan V, Dutt S, Manjappa R, Lau B, Soto LA, Ashraf MR, Skinner L, Yu SJ, Surucu M, Casey KM, Rankin EB, Graves E, Lu W, Loo BW, Gu X. Exploring Deep Learning for Estimating the Isoeffective Dose of FLASH Irradiation From Mouse Intestinal Histological Images. Int J Radiat Oncol Biol Phys 2024; 119:1001-1010. [PMID: 38171387 DOI: 10.1016/j.ijrobp.2023.12.032] [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: 07/05/2023] [Revised: 12/09/2023] [Accepted: 12/23/2023] [Indexed: 01/05/2024]
Abstract
PURPOSE Ultrahigh-dose-rate (FLASH) irradiation has been reported to reduce normal tissue damage compared with conventional dose rate (CONV) irradiation without compromising tumor control. This proof-of-concept study aims to develop a deep learning (DL) approach to quantify the FLASH isoeffective dose (dose of CONV that would be required to produce the same effect as the given physical FLASH dose) with postirradiation mouse intestinal histology images. METHODS AND MATERIALS Eighty-four healthy C57BL/6J female mice underwent 16 MeV electron CONV (0.12 Gy/s; n = 41) or FLASH (200 Gy/s; n = 43) single fraction whole abdominal irradiation. Physical dose ranged from 12 to 16 Gy for FLASH and 11 to 15 Gy for CONV in 1 Gy increments. Four days after irradiation, 9 jejunum cross-sections from each mouse were hematoxylin and eosin stained and digitized for histological analysis. CONV data set was randomly split into training (n = 33) and testing (n = 8) data sets. ResNet101-based DL models were retrained using the CONV training data set to estimate the dose based on histological features. The classical manual crypt counting (CC) approach was implemented for model comparison. Cross-section-wise mean squared error was computed to evaluate the dose estimation accuracy of both approaches. The validated DL model was applied to the FLASH data set to map the physical FLASH dose into the isoeffective dose. RESULTS The DL model achieved a cross-section-wise mean squared error of 0.20 Gy2 on the CONV testing data set compared with 0.40 Gy2 of the CC approach. Isoeffective doses estimated by the DL model for FLASH doses of 12, 13, 14, 15, and 16 Gy were 12.19 ± 0.46, 12.54 ± 0.37, 12.69 ± 0.26, 12.84 ± 0.26, and 13.03 ± 0.28 Gy, respectively. CONCLUSIONS Our proposed DL model achieved accurate CONV dose estimation. The DL model results indicate that in the physical dose range of 13 to 16 Gy, the biologic dose response of small intestinal tissue to FLASH irradiation is represented by a lower isoeffective dose compared with the physical dose. Our DL approach can be a tool for studying isoeffective doses of other radiation dose modifying interventions.
Collapse
Affiliation(s)
- Jie Fu
- Department of Radiation Oncology, University of Washington, Seattle, Washington; Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Zi Yang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California; Medical Artificial Intelligence and Automation (MAIA) Laboratory, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Stavros Melemenidis
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Vignesh Viswanathan
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Suparna Dutt
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Rakesh Manjappa
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Brianna Lau
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Luis A Soto
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - M Ramish Ashraf
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Lawrie Skinner
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Shu-Jung Yu
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Murat Surucu
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Kerriann M Casey
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, California
| | - Erinn B Rankin
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Edward Graves
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Weiguo Lu
- Medical Artificial Intelligence and Automation (MAIA) Laboratory, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Billy W Loo
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California.
| | - Xuejun Gu
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|