1
|
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; 122:443-457. [PMID: 39848295 DOI: 10.1016/j.ijrobp.2025.01.006] [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: 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
|
2
|
Battestini M, Missiaggia M, Bolzoni S, Cordoni FG, Scifoni E. A multiscale radiation biophysical stochastic model describing the cell survival response at ultra-high dose rate under different oxygenations and radiation qualities. Radiother Oncol 2025; 207:110895. [PMID: 40233874 DOI: 10.1016/j.radonc.2025.110895] [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: 01/12/2025] [Revised: 03/25/2025] [Accepted: 04/09/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND AND PURPOSE While the advantages of ultra-high dose-rate (UHDR) irradiation have been well highlighted experimentally, the biological mechanism underlying the FLASH effect is still unclear and highly debated. The aim of this work is to reproduce the main in-vitro UHDR experiments and to try to explain the different in-vivo response between healthy tissues and tumors, developing a fully consistent radiation biophysical model for UHDR regime. MATERIALS AND METHODS We developed the MultiScale Generalized Stochastic Microdosimetric Model (MS-GSM2), a multi-stage extension of the GSM2, which is a probabilistic model describing the time evolution of the lesions in an irradiated cell nucleus. We coupled the slow DNA damage evolution with the fast chemical reaction kinetics, including the impact of the redox environment. RESULTS The MS-GSM2 can investigate the combined effects of chemical species, DNA damage formation and time evolution. We demonstrate that the MS-GSM2 predictions are coherent with the in-vitro UHDR experimental results across various oxygenation levels, and radiation qualities. We analyze the role of the chemical environmental conditions of the irradiated medium, i.e. oxygenation, and scavengers concentration, discussing possible factors that can attenuate or level out the dose rate dependence of the cell survival, to understand the differential effect that occurs in-vivo between normal tissue and tumor. CONCLUSION The MS-GSM2 can accurately describe multiple aspects of the FLASH effect and be consistent with the main evidence from the in-vitro experiments with different types of radiation and oxygenations. Our model proposes a consistent explanation for the differential outcomes observed in normal tissues and tumors, in-vivo and in-vitro.
Collapse
Affiliation(s)
- Marco Battestini
- Department of Physics, University of Trento 38123 Trento, Italy; Trento Institute for Fundamental Physics and Applications (TIFPA), National Institute for Nuclear Physics (INFN), 38123 Trento, Italy
| | - Marta Missiaggia
- Trento Institute for Fundamental Physics and Applications (TIFPA), National Institute for Nuclear Physics (INFN), 38123 Trento, Italy; Radiation Oncology Department, University of Miami FL, 33136 Miami, USA
| | - Sara Bolzoni
- Department of Physics, University of Trento 38123 Trento, Italy
| | - Francesco G Cordoni
- Trento Institute for Fundamental Physics and Applications (TIFPA), National Institute for Nuclear Physics (INFN), 38123 Trento, Italy; Department of Civil, Environmental and Mechanical Engineering, University of Trento 38123 Trento, Italy.
| | - Emanuele Scifoni
- Trento Institute for Fundamental Physics and Applications (TIFPA), National Institute for Nuclear Physics (INFN), 38123 Trento, Italy
| |
Collapse
|
3
|
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; 122:491-501. [PMID: 39862897 DOI: 10.1016/j.ijrobp.2025.01.007] [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: 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
|
4
|
Hörberger F, Petersson K, Ceberg S, Bäck S, Adrian G, Ceberg C. Investigating the therapeutic potential of FLASH radiotherapy - a treatment planning study. Radiother Oncol 2025:110947. [PMID: 40409370 DOI: 10.1016/j.radonc.2025.110947] [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: 01/24/2025] [Revised: 04/30/2025] [Accepted: 05/16/2025] [Indexed: 05/25/2025]
Abstract
PURPOSE/BACKGROUND Ultra-high dose rate radiotherapy (RT) has shown potential for differential normal tissue (NT) sparing (a phenomenon termed the "FLASH effect"), particularly for larger fraction doses (>5 Gy). However, transitioning to hypofractionation may increase late-reacting NT toxicity, counteracting the FLASH effect. This study evaluates whether FLASH-RT can provide netsparing for organs at risk (OARs) and NT within the PTV under the assumption of standard-of-care dose-conformity. MATERIAL/METHODS Five patients per tumor-site (breast, head-and-neck, prostate, and glioblastoma) were analyzed. Using the Linear-Quadratic model, dose-distributions with higher dose per fraction were derived from standard schedules while maintaining tumor control efficacy. FLASH-modified dose-distributions were simulated voxel-by-voxel using logistic regression-based dose-modifying factors modeled from preclinical data. These plans were converted to standard fractionation equivalents for radiobiological comparisons of NT damage. Netsparing was defined as the difference in OAR dose-volume histogram parameters between standard and FLASH-modified plans, normalized to the prescribed dose. Commonly used α/β-ratios for tumors and late-reacting NT were applied. RESULTS The netsparing for OARs and PTV varied strongly by tumor location. Breast and prostate cases showed positive netsparing, indicating that the FLASH effect outweighed increased toxicity. Even under a conservative scenario (higher α/βT vs. α/βNT), most OARs showed positive netsparing. In glioblastoma and head-and-neck cases, no netsparing was observed, indicating increased toxicity even with FLASH induced NT-sparing. CONCLUSION FLASH-RT appears to be beneficial for tumor sites where α/βT ≲ α/βNT, such as breast and prostate. However, not all tumor sites may benefit from FLASH-RT, highlighting the need for site-specific consideration for FLASH-RT implementation.
Collapse
Affiliation(s)
- Filip Hörberger
- Medical Radiation Physics, Department of Clinical Sciences Lund, Lund University, Lund, Sweden.
| | - Kristoffer Petersson
- Radiation Physics, Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Sweden; Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Sofie Ceberg
- Medical Radiation Physics, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Sven Bäck
- Radiation Physics, Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Sweden
| | - Gabriel Adrian
- Radiation Physics, Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Sweden; Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Crister Ceberg
- Medical Radiation Physics, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| |
Collapse
|
5
|
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
|
6
|
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; 206:110796. [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] [MESH Headings] [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, Aarhus, 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 Graversen 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, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
7
|
Wei S, Lin H, Cheng C, Choi JI, Simone CB, Kang M. An ultra-high dose rate Bragg peak tracking technique provides more affordable proton radiotherapy for cancer patients: From principle to experimental validation. Radiother Oncol 2025; 206:110800. [PMID: 39988304 DOI: 10.1016/j.radonc.2025.110800] [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: 08/07/2024] [Revised: 02/11/2025] [Accepted: 02/14/2025] [Indexed: 02/25/2025]
Abstract
PURPOSE This work aims to experimentally validate a novel cost-effective solution for achieving both conventional dose-rate and ultra-high dose rate (UHDR) deliveries in pencil beam scanning proton therapy. METHODS A proton therapy delivery solution was previously developed by our group using only a single pristine Bragg peak of the highest energy proton beams from a cyclotron. This approach streamlines upstream beam modifiers, including energy degrader, selection and focusing systems, while utilizing of universal range shifters (URS) and range compensators (RCs) to preserve high beam transmission efficiency for UHDR beam delivery. It achieves the Bragg peak tracking and target dose conformity, making it potentially suitable for FLASH radiation therapy. In the current study, we highlighted the realization of the solution by using URS and customized beam-specific RCs via simulation in an in-house treatment planning software (TPS) which is then fabricated by a 3D printer, facilitating precise beam shaping and Bragg peak tracking. Experimental validation of this method was conducted using a clinical proton system to showcase a practical solution that can be translated into realistic operation. Both dose and dose rate were measured and compared to treatment planning results. RESULTS The proton convolution superposition (PCS) dose calculation was benchmarked by the Monte Carlo calculation. Matrixx PT measured the delivered dose in the uniform and head-neck (HN) phantom, and the gamma passing rates were > 99 % in the water phantom. The gamma rate was > 98 % for the HN phantom for this distal tracking method. The measured dose difference between the TPS and HN phantom was < 2 %. The implementation of a high temporal resolution strip ion chamber detector array enabled accurate measurement of the spot time structure, facilitating 3D dose rate reconstruction across various beam currents. CONCLUSION The experimental validation successfully demonstrated the dosimetric accuracy and robustness of this proposed delivery method. The employment of the Bragg peak tracking method holds great promise for reducing treatment delivery costs for future UHDR and conventional dose rate proton radiation therapy, ultimately benefiting a larger population of patients.
Collapse
Affiliation(s)
- Shouyi Wei
- New York Proton Center, New York, NY 10035, USA.
| | - Haibo Lin
- New York Proton Center, New York, NY 10035, USA.
| | - Chingyun Cheng
- Department of Human Oncology, University of Wisconsin, Madison, Wisconsin 53792, USA.
| | | | | | - Minglei Kang
- New York Proton Center, New York, NY 10035, USA.
| |
Collapse
|
8
|
Toschini M, Colizzi I, Lomax AJ, Psoroulas S. Medical physics dataset article: A database of FLASH murine in vivo studies. Med Phys 2025. [PMID: 40270058 DOI: 10.1002/mp.17744] [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: 10/12/2024] [Revised: 01/15/2025] [Accepted: 02/25/2025] [Indexed: 04/25/2025] Open
Abstract
PURPOSE The FLASH effect refers to a lower normal tissue damage for an equivalent tumor response, potentially widening the therapeutic window for radiotherapy. Although this effect has been demonstrated in various experiments using different types of particles and irradiation parameters, the underlying mechanism is not yet clearly understood. Uncertainties surround the conducted experiments, the explored parameter space, and the variability of reported results. To gain a better overview, we have created a dataset that includes in vivo FLASH experiments. This dataset documents all machine and biological dosimetric parameters, and for determined endpoints, it includes the outcome of the experiment. Our goal with this database is to increase awareness of the results and their variability and provide a useful research and analysis tool for the community. ACQUISITION AND VALIDATION METHODS The database contains peer-reviewed papers published until March 2024 on the FLASH in vivo (murine) experiments. From each paper, previously defined parameters have been manually extracted and/or recalculated to ensure compatibility within the database entries. DATA FORMAT AND USAGE NOTES We provide two types of datasets: a user-friendly web-based Notion database and spreadsheets on a Zenodo repository. The database contains all the reviewed papers with extracted information in text or numeric form. Users can duplicate the database or view, search, filter, and reorganize online entries. The spreadsheets contain the data for the most analyzed endpoints (skin toxicity, survival rate, and crypt cells), allowing a comparative analysis. POTENTIAL APPLICATIONS The study has two main applications. The web-based database will allow for a user-friendly search of information and metadata of all published FLASH murine data. This will facilitate future research efforts to better understand the FLASH effect. The spreadsheets are a simple and useful tool for the community to conduct statistical analysis and determine the parameters associated with the FLASH effect.
Collapse
Affiliation(s)
- Mathilde Toschini
- Center for Proton Therapy, Paul Scherrer Institut, Villigen PSI, Switzerland
- Department of Physics, ETH Zürich, Zürich, Switzerland
| | - Isabella Colizzi
- Center for Proton Therapy, Paul Scherrer Institut, Villigen PSI, Switzerland
- Department of Physics, ETH Zürich, Zürich, Switzerland
| | - Antony John Lomax
- Center for Proton Therapy, Paul Scherrer Institut, Villigen PSI, Switzerland
- Department of Physics, ETH Zürich, Zürich, Switzerland
| | - Serena Psoroulas
- Center for Proton Therapy, Paul Scherrer Institut, Villigen PSI, Switzerland
- Department of Radiation Oncology, University Hospital Zurich (USZ), University of Zurich (UZH), Zurich, Switzerland
| |
Collapse
|
9
|
Manring HR, Fleming JL, Meng W, Gamez ME, Blakaj DM, Chakravarti A. FLASH Radiotherapy: From In Vivo Data to Clinical Translation. Hematol Oncol Clin North Am 2025; 39:237-255. [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] [MESH Headings] [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 E 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
|
10
|
Poulsen PR, Johansen JG, Sitarz MK, Kanouta E, Kristensen L, Grau C, Sørensen BS. In Reply to Scifoni et al. Int J Radiat Oncol Biol Phys 2025; 121:1400-1401. [PMID: 40089344 DOI: 10.1016/j.ijrobp.2024.12.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 12/22/2024] [Indexed: 03/17/2025]
Affiliation(s)
- Per Rugaard Poulsen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Denmark.
| | - Jacob Graversen Johansen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Denmark
| | - Mateusz Krzysztof Sitarz
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Denmark
| | - Eleni Kanouta
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Denmark
| | - Line Kristensen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Denmark; Department of Experimental Clinical Oncology, Aarhus University Hospital, Denmark
| | - Cai Grau
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Denmark
| | - Brita Singers Sørensen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Denmark; Department of Experimental Clinical Oncology, Aarhus University Hospital, Denmark
| |
Collapse
|
11
|
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] [MESH Headings] [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
|
12
|
Klaver YLB, Hoogeman MS, Lu QR, Bradley JD, Choi JI, Ferris MJ, Grau C, Guha C, Lin H, Lin L, Mascia AE, Moerman AM, Poulsen PR, Shi LZ, Singers Sørensen B, Tian S, Vozenin MC, Willey CD, Zhou S, Amos RA, Hawkins M, Simone CB. Requirements and Study Design for the Next Proton FLASH Clinical Trials: an International Multidisciplinary Delphi Consensus. Int J Radiat Oncol Biol Phys 2025:S0360-3016(25)00306-2. [PMID: 40174648 DOI: 10.1016/j.ijrobp.2025.03.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 02/20/2025] [Accepted: 03/22/2025] [Indexed: 04/04/2025]
Abstract
PURPOSE The FLASH effect, defined as normal tissue sparing while maintaining tumor control with ultra-high dose-rate irradiation, has been demonstrated preclinically in different tumors and tissues. Although the biological mechanisms are unclear, there is a need for clinical trials investigating the value of proton FLASH irradiation (pFLASH). The purpose of this study was to establish an expert consensus regarding prerequisites, study design, and endpoints for the next clinical trials exploring the clinical potentials of pFLASH. METHODS AND MATERIALS Delphi methodology was used to develop a systematic expert consensus. An international expert panel was composed of 21 clinicians, physicists, and biologists, well-balanced in expertise and geography, using predefined inclusion criteria. Statements were scored on a 5-point Likert scale in 2 rounds of online questionnaire voting. The definition of consensus was set a priori. RESULTS The response rate was 100% in both rounds. Preclinical in vivo demonstration of the FLASH effect in normal tissue while maintaining tumor response was deemed essential before starting a clinical trial in a specific tumor site. The next clinical pFLASH trials are advised to include adult patients only, with a minimal expected overall survival of 1 year for palliative settings or, preferably, oligometastatic disease in the ablative setting. The pFLASH effect should be studied in a single treatment modality setting with toxicity reduction as the primary endpoint. Recommendations were formulated on the use of clinical targets and organs at risk constraints, requirements for evaluation and reporting, and accuracy levels and pretreatment verification of dose rates. No consensus was reached on the use of multiple beams, multiple fractions, and fraction dose. CONCLUSIONS There is a need for additional data regarding the influence of fractionation and multiple beam planning. The results of this study can be used to develop roadmaps to guide future clinical trial design.
Collapse
Affiliation(s)
- Yvonne L B Klaver
- HollandPTC, Delft, The Netherlands; Department of Radiotherapy, Leiden University Medical Center, Leiden, The Netherlands.
| | - Mischa S Hoogeman
- HollandPTC, Delft, The Netherlands; Department of Radiotherapy, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Q Richard Lu
- Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jeffrey D Bradley
- Department of Radiation Oncology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - J Isabelle Choi
- Department of Radiation Oncology, New York Proton Center, New York, New York; Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Matthew J Ferris
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland; Maryland Proton Treatment Center, Baltimore, Maryland
| | - Cai Grau
- Danish Center for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
| | - Chandan Guha
- Department of Radiation Oncology, Montefiore University Hospital, Bronx, New York
| | - Haibo Lin
- Department of Radiation Oncology, New York Proton Center, New York, New York; Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Radiation Oncology, Montefiore University Hospital, Bronx, New York
| | - Liyong Lin
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Anthony E Mascia
- Cincinnati Children's Hospital Medical Center, Cancer and Blood Disease Institute, Division of Oncology, Cincinnati, Ohio
| | | | - Per R Poulsen
- Danish Center for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Lewis Z Shi
- Departments of Microbiology, Pharmacology & Toxicology; The Immunology Institute; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama; Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Brita Singers Sørensen
- Danish Center for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Experimental Clinical Oncology, Aarhus University, Denmark
| | - Sibo Tian
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Marie-Catherine Vozenin
- Radiotherapy and Radiobiology Sector, Radiation Therapy Service, University Hospital of Geneva, Geneva, Switzerland
| | - Christopher D Willey
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Sumin Zhou
- Radiation Oncology Department, University of Nebraska Medical Center, Omaha, Nebraska
| | - Richard A Amos
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Maria Hawkins
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom; Clinical Oncology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Charles B Simone
- Department of Radiation Oncology, New York Proton Center, New York, New York; Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
13
|
Williams MT, Regan SL, Fritz AL, Gollaway BM, Mascia AE, Vatner RE, Perentesis JP, Vorhees CV. Effects of whole brain proton irradiation at conventional or ultra-high dose rate (FLASH), in adult male Sprague Dawley rats. Sci Rep 2025; 15:10602. [PMID: 40148391 PMCID: PMC11950509 DOI: 10.1038/s41598-025-94534-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
Radiation is an effective treatment for many brain tumors, but often causes cognitive impairment. Ultra-high dose rate (FLASH) radiotherapy is less toxic to many normal tissues and may protect against adverse cognitive effects of cranial irradiation. Adult male Sprague Dawley rats received a single 18 Gy fraction of cranial irradiation with protons at 1 Gy/s (CV), 60 Gy/s (FLASH-60), 95 Gy/s (FLASH-95), or sham treatment (Control) (n ≥ 22/group). Rats were tested in open-field, acoustic (ASR) and tactile startle (TSR), novelty preference, radial water maze (RWM), Morris water maze (MWM), Cincinnati water maze configurations A and B (CWM-A CWM-B), and novelty tests. Locomotion was decreased and TSR increased in all irradiated rats and ASR increased in FLASH-95 rats compared with Controls. In MWM acquisition and reversal, the CV and FLASH-60 rats had reduced path efficiency but during shift and shift reversal all irradiated rats had increased latencies and reduced path efficiencies compared with Controls. In CWM-A all irradiated rats performed below Controls. There were no differences found in CWM-B, novelty tests, or RWM. In summary, FLASH treatment after 18 Gy cranial proton irradiation did not result in reduced cognitive toxicity.
Collapse
Affiliation(s)
- Michael T Williams
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA.
- Division of Neurology (MLC 7044), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229-3039, USA.
- Cincinnati Children's/University of Cincinnati Proton Therapy and Research Center, Cincinnati, OH, 45229, USA.
| | - Samantha L Regan
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
- Division of Neurology (MLC 7044), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229-3039, USA
- Department of Human Genetics, University of Michigan Medical Center, 3703 E. Catherine St., Ann Arbor, MI, 48109-5618, USA
| | - Adam L Fritz
- Division of Neurology (MLC 7044), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229-3039, USA
| | - Brooke M Gollaway
- Division of Neurology (MLC 7044), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229-3039, USA
| | - Anthony E Mascia
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
- Cincinnati Children's/University of Cincinnati Proton Therapy and Research Center, Cincinnati, OH, 45229, USA
- Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Ralph E Vatner
- Cincinnati Children's/University of Cincinnati Proton Therapy and Research Center, Cincinnati, OH, 45229, USA
- Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - John P Perentesis
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
- Cincinnati Children's/University of Cincinnati Proton Therapy and Research Center, Cincinnati, OH, 45229, USA
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Charles V Vorhees
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
- Division of Neurology (MLC 7044), Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH, 45229-3039, USA
- Cincinnati Children's/University of Cincinnati Proton Therapy and Research Center, Cincinnati, OH, 45229, USA
| |
Collapse
|
14
|
Melemenidis S, Viswanathan V, Dutt S, Kapadia N, Lau B, Soto LA, Ashraf MR, Thakur B, Mutahar AZI, Skinner LB, Yu AS, Surucu M, Casey KM, Rankin EB, Horst KC, Graves EE, Loo BW, Dirbas FM. Effectiveness of FLASH vs. Conventional Dose Rate Radiotherapy in a Model of Orthotopic, Murine Breast Cancer. Cancers (Basel) 2025; 17:1095. [PMID: 40227580 PMCID: PMC11988084 DOI: 10.3390/cancers17071095] [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: 02/05/2025] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 04/15/2025] Open
Abstract
Introduction: Radiotherapy is effective for breast cancer treatment but often causes undesirable side effects that impair quality of life. Ultra-high dose rate radiotherapy (FLASH) has shown reduced normal tissue toxicity while achieving comparable tumor growth delay compared to conventional dose rate radiotherapy (CONV). This study evaluated whether FLASH could achieve similar tumor control as CONV with tumor eradication as the primary endpoint, in an orthotopic breast cancer model. Methods: Non-metastatic, orthotopic tumors were generated in the left fourth mammary fat pad using the Py117 mammary tumor cell line in syngeneic C57BL/6J mice. Two sequential irradiation studies were performed using FLASH (93-200 Gy/s) and CONV (0.08 Gy/s) electron beams. Single fractions of 20, 25, or 30 Gy were applied to tumors with varying abdominal wall treatment fields (~3.75 or 2.5 mm treatment margin to tumor). Results: Both FLASH and CONV demonstrated comparable efficacy. Small tumors treated with 30 Gy and larger abdominal wall treatment fields appeared to have complete eradication at 30 days but also exhibited the highest skin toxicity, limiting follow-up and preventing confirmation of eradication. Smaller abdominal wall treatment fields reduced skin toxicity and allowed for extended follow-up, which resulted in 75% tumor-free survival at 48 days. Larger tumors showed growth delay but no eradication. Conclusions: In this preclinical, non-metastatic orthotopic breast cancer model, FLASH and CONV demonstrated equivalent tumor control with single-fraction doses of 20, 25, or 30 Gy. Overall, 30 Gy achieved the highest eradication rate but also resulted in the most pronounced skin toxicity.
Collapse
Affiliation(s)
- Stavros Melemenidis
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; (S.M.); (V.V.); (S.D.); (B.L.); (L.A.S.); (M.R.A.); (L.B.S.); (A.S.Y.); (M.S.); (E.B.R.); (K.C.H.); (E.E.G.); (B.W.L.J.)
| | - Vignesh Viswanathan
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; (S.M.); (V.V.); (S.D.); (B.L.); (L.A.S.); (M.R.A.); (L.B.S.); (A.S.Y.); (M.S.); (E.B.R.); (K.C.H.); (E.E.G.); (B.W.L.J.)
| | - Suparna Dutt
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; (S.M.); (V.V.); (S.D.); (B.L.); (L.A.S.); (M.R.A.); (L.B.S.); (A.S.Y.); (M.S.); (E.B.R.); (K.C.H.); (E.E.G.); (B.W.L.J.)
| | - Naviya Kapadia
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; (S.M.); (V.V.); (S.D.); (B.L.); (L.A.S.); (M.R.A.); (L.B.S.); (A.S.Y.); (M.S.); (E.B.R.); (K.C.H.); (E.E.G.); (B.W.L.J.)
| | - Brianna Lau
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; (S.M.); (V.V.); (S.D.); (B.L.); (L.A.S.); (M.R.A.); (L.B.S.); (A.S.Y.); (M.S.); (E.B.R.); (K.C.H.); (E.E.G.); (B.W.L.J.)
| | - Luis A. Soto
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; (S.M.); (V.V.); (S.D.); (B.L.); (L.A.S.); (M.R.A.); (L.B.S.); (A.S.Y.); (M.S.); (E.B.R.); (K.C.H.); (E.E.G.); (B.W.L.J.)
| | - M. Ramish Ashraf
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; (S.M.); (V.V.); (S.D.); (B.L.); (L.A.S.); (M.R.A.); (L.B.S.); (A.S.Y.); (M.S.); (E.B.R.); (K.C.H.); (E.E.G.); (B.W.L.J.)
| | - Banita Thakur
- Department of Surgery, Stanford University School of Medicine, Stanford Cancer Institute, Stanford, CA 94305, USA; (B.T.); (A.Z.I.M.)
| | - Adel Z. I. Mutahar
- Department of Surgery, Stanford University School of Medicine, Stanford Cancer Institute, Stanford, CA 94305, USA; (B.T.); (A.Z.I.M.)
| | - Lawrie B. Skinner
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; (S.M.); (V.V.); (S.D.); (B.L.); (L.A.S.); (M.R.A.); (L.B.S.); (A.S.Y.); (M.S.); (E.B.R.); (K.C.H.); (E.E.G.); (B.W.L.J.)
| | - Amy S. Yu
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; (S.M.); (V.V.); (S.D.); (B.L.); (L.A.S.); (M.R.A.); (L.B.S.); (A.S.Y.); (M.S.); (E.B.R.); (K.C.H.); (E.E.G.); (B.W.L.J.)
| | - Murat Surucu
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; (S.M.); (V.V.); (S.D.); (B.L.); (L.A.S.); (M.R.A.); (L.B.S.); (A.S.Y.); (M.S.); (E.B.R.); (K.C.H.); (E.E.G.); (B.W.L.J.)
| | - Kerriann M. Casey
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA;
| | - Erinn B. Rankin
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; (S.M.); (V.V.); (S.D.); (B.L.); (L.A.S.); (M.R.A.); (L.B.S.); (A.S.Y.); (M.S.); (E.B.R.); (K.C.H.); (E.E.G.); (B.W.L.J.)
| | - Kathleen C. Horst
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; (S.M.); (V.V.); (S.D.); (B.L.); (L.A.S.); (M.R.A.); (L.B.S.); (A.S.Y.); (M.S.); (E.B.R.); (K.C.H.); (E.E.G.); (B.W.L.J.)
| | - Edward E. Graves
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; (S.M.); (V.V.); (S.D.); (B.L.); (L.A.S.); (M.R.A.); (L.B.S.); (A.S.Y.); (M.S.); (E.B.R.); (K.C.H.); (E.E.G.); (B.W.L.J.)
| | - Billy W. Loo
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA; (S.M.); (V.V.); (S.D.); (B.L.); (L.A.S.); (M.R.A.); (L.B.S.); (A.S.Y.); (M.S.); (E.B.R.); (K.C.H.); (E.E.G.); (B.W.L.J.)
| | - Frederick M. Dirbas
- Department of Surgery, Stanford University School of Medicine, Stanford Cancer Institute, Stanford, CA 94305, USA; (B.T.); (A.Z.I.M.)
| |
Collapse
|
15
|
Chaoui M, Bouhali O, Tayalati Y. FLASH radiotherapy: technical advances, evidence of the FLASH effect and mechanistic insights. Biomed Phys Eng Express 2025; 11:022003. [PMID: 40043321 DOI: 10.1088/2057-1976/adbcb1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 03/05/2025] [Indexed: 03/18/2025]
Abstract
Cancer is one of the leading causes of death worldwide, responsible for nearly 10 million deaths in 2020, with approximately 50% of patients receiving radiation therapy as part of their treatment (Baskaret al2012). Preclinical investigations studies have shown that FLASH radiotherapy (FLASH-RT), delivering radiation in ultra-high dose rates (UHDR), preserves healthy tissue integrity and reduces toxicity, all while maintaining an effective tumor response compared to conventional radiotherapy (CONV-RT), the combined biological benefit was termed as FLASH effect. This article comprehensively surveys pertinent research conducted within FLASH-RT, explores the facilities used in this realm, delves into hypothesized mechanism perspectives, and addresses the challenges to trigger the FLASH effect. In addition, we discuss the potential prospects of FLASH-RT and examine the obstacles that require resolution before its clinical implementation can become a reality.
Collapse
Affiliation(s)
- Mustapha Chaoui
- Faculty of Sciences, University Mohammed V in Rabat, Morocco
| | - Othmane Bouhali
- Electrical Engineering, College of Science and Engineering, Hamad Bin Khalifa University Doha, Qatar
| | - Yahya Tayalati
- Faculty of Sciences, University Mohammed V in Rabat, Morocco
- Institute of Applied Physics, Mohammed VI Polytechnic University, Ben Guerir, Morocco
| |
Collapse
|
16
|
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
|
17
|
Akulinichev SV, Glukhov SI, Kuznetsova EA, Gavrilov YK, Kokontsev DA, Martynova VV, Merzlikin GV, Yakovlev IA. Manifestation of the FLASH effect in proton irradiation of embryos. Int J Radiat Biol 2024; 101:144-152. [PMID: 39625863 DOI: 10.1080/09553002.2024.2435338] [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: 08/08/2024] [Revised: 10/30/2024] [Accepted: 11/24/2024] [Indexed: 01/25/2025]
Abstract
PURPOSE In order to study the FLASH effect using live models, this work compared proton-induced damage to embryos (nine days after fertilization) and one-day-old chicks (18 days after fertilization) from irradiated at different dose rates eggs of Japanese quail (Coturnix coturnix japónica). MATERIALS AND METHODS Eggs were irradiated with protons in different modes depending on the dose rate: in a conventional mode (<1 Gy/s, CONV), in a flash mode (∼100 Gy/s, FLASH) and in a single-pulse flash mode (∼105 Gy/s SPLASH). RESULTS By the criteria of body weight and length, as well as the number of erythrocytes with micronuclei in nine-day-old embryos from eggs irradiated in the spread-out Bragg peak (SOBP) (8.5 Gy), FLASH and SPLASH modes were found to be less traumatic compared with the CONV mode. Among all irradiated embryos, the maximum body weight and length were observed in the SPLASH mode. The lowest death incidence and the smallest number of abnormal erythrocytes were recorded after FLASH and SPLASH irradiation. In chicks that hatched from eggs irradiated in the CONV mode, a tendency for an increase in the number of abnormal erythrocytes was observed. The speed of movement of chicks from FLASH- and SPLASH-irradiated eggs was comparable with that from unirradiated eggs, while chicks from eggs irradiated in the CONV mode were less active than all others. CONCLUSIONS The proton irradiation of eggs in SOBP using high dose-rate modes is less damaging for healthy tissues and for the development of embryos and chicks on the cellular, anatomical, and physiological levels.
Collapse
Affiliation(s)
- S V Akulinichev
- Institute for Nuclear Research, Russian Academy of Sciences (INR RAS), Troitsk, Russia
- B.V.Petrovsky National Research Center of Surgery, Moscow, Russia
| | - S I Glukhov
- Institute for Nuclear Research, Russian Academy of Sciences (INR RAS), Troitsk, Russia
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences (ITEB RAS), Pushchino, Russia
| | - E A Kuznetsova
- Institute for Nuclear Research, Russian Academy of Sciences (INR RAS), Troitsk, Russia
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences (ITEB RAS), Pushchino, Russia
| | - Yu K Gavrilov
- Institute for Nuclear Research, Russian Academy of Sciences (INR RAS), Troitsk, Russia
| | - D A Kokontsev
- Institute for Nuclear Research, Russian Academy of Sciences (INR RAS), Troitsk, Russia
| | - V V Martynova
- Institute for Nuclear Research, Russian Academy of Sciences (INR RAS), Troitsk, Russia
| | - G V Merzlikin
- Institute for Nuclear Research, Russian Academy of Sciences (INR RAS), Troitsk, Russia
| | - I A Yakovlev
- Institute for Nuclear Research, Russian Academy of Sciences (INR RAS), Troitsk, Russia
- B.V.Petrovsky National Research Center of Surgery, Moscow, Russia
| |
Collapse
|
18
|
Wang J, Melemenidis S, Manjappa R, Viswanathan V, Ashraf RM, Levy K, Skinner LB, Soto LA, Chow S, Lau B, Ko RB, Graves EE, Yu AS, Bush KK, Surucu M, Rankin EB, Loo BW, Schüler E, Maxim PG. Dosimetric calibration of anatomy-specific ultra-high dose rate electron irradiation platform for preclinical FLASH radiobiology experiments. Med Phys 2024; 51:9166-9178. [PMID: 39331834 DOI: 10.1002/mp.17432] [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: 03/14/2024] [Revised: 07/27/2024] [Accepted: 09/06/2024] [Indexed: 09/29/2024] Open
Abstract
BACKGROUND FLASH radiation therapy (RT) offers a promising avenue for the broadening of the therapeutic index. However, to leverage the full potential of FLASH in the clinical setting, an improved understanding of the biological principles involved is critical. This requires the availability of specialized equipment optimized for the delivery of conventional (CONV) and ultra-high dose rate (UHDR) irradiation for preclinical studies. One method to conduct such preclinical radiobiological research involves adapting a clinical linear accelerator configured to deliver both CONV and UHDR irradiation. PURPOSE We characterized the dosimetric properties of a clinical linear accelerator configured to deliver ultra-high dose rate irradiation to two anatomic sites in mice and for cell-culture FLASH radiobiology experiments. METHODS Delivered doses of UHDR electron beams were controlled by a microcontroller and relay interfaced with the respiratory gating system. We also produced beam collimators with indexed stereotactic mouse positioning devices to provide anatomically specific preclinical treatments. Treatment delivery was monitored directly with an ionization chamber, and charge measurements were correlated with radiochromic film measurements at the entry surface of the mice. The setup for conventional dose rate irradiation utilized the same collimation system but at increased source-to-surface distance. Monte Carlo simulations and film dosimetry were used to characterize beam properties and dose distributions. RESULTS The mean electron beam energies before the flattening filter were 18.8 MeV (UHDR) and 17.7 MeV (CONV), with corresponding values at the mouse surface of 17.2 and 16.2 MeV. The charges measured with an external ion chamber were linearly correlated with the mouse entrance dose. The use of relay gating for pulse control initially led to a delivery failure rate of 20% (± 1 pulse); adjustments to account for the linac latency improved this rate to < 1/20. Beam field sizes for two anatomically specific mouse collimators (4 × 4 cm2 for whole-abdomen and 1.5 × 1.5 cm2 for unilateral lung irradiation) were accurate within < 5% and had low radiation leakage (< 4%). Normalizing the dose at the center of the mouse (∼0.75 cm depth) produced UHDR and CONV doses to the irradiated volumes with > 95% agreement. CONCLUSION We successfully configured a clinical linear accelerator for increased output and developed a robust preclinical platform for anatomically specific irradiation, with highly accurate and precise temporal and spatial dose delivery, for both CONV and UHDR irradiation applications.
Collapse
Affiliation(s)
- Jinghui Wang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Stavros Melemenidis
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Rakesh Manjappa
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Vignesh Viswanathan
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Ramish M Ashraf
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Karen Levy
- Department of Gynecologic Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Lawrie B Skinner
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Luis A Soto
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Stephanie Chow
- Department of Gynecologic Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Brianna Lau
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Ryan B Ko
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Edward E Graves
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Amy S Yu
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Karl K Bush
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Murat Surucu
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Erinn B Rankin
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
- Department of Gynecologic Oncology, Stanford University School of Medicine, Stanford, California, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Billy W Loo
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Emil Schüler
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Peter G Maxim
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
19
|
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
|
20
|
Malimban J, Ludwig F, Lathouwers D, Staring M, Verhaegen F, Brandenburg S. A simulation framework for preclinical proton irradiation workflow. Phys Med Biol 2024; 69:215040. [PMID: 39433066 DOI: 10.1088/1361-6560/ad897f] [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: 06/27/2024] [Accepted: 10/21/2024] [Indexed: 10/23/2024]
Abstract
Objective.The integration of proton beamlines with x-ray imaging/irradiation platforms has opened up possibilities for image-guided Bragg peak irradiations in small animals. Such irradiations allow selective targeting of normal tissue substructures and tumours. However, their small size and location pose challenges in designing experiments. This work presents a simulation framework useful for optimizing beamlines, imaging protocols, and design of animal experiments. The usage of the framework is demonstrated, mainly focusing on the imaging part.Approach.The fastCAT toolkit was modified with Monte Carlo (MC)-calculated primary and scatter data of a small animal imager for the simulation of micro-CT scans. The simulated CT of a mini-calibration phantom from fastCAT was validated against a full MC TOPAS CT simulation. A realistic beam model of a preclinical proton facility was obtained from beam transport simulations to create irradiation plans in matRad. Simulated CT images of a digital mouse phantom were generated using single-energy CT (SECT) and dual-energy CT (DECT) protocols and their accuracy in proton stopping power ratio (SPR) estimation and their impact on calculated proton dose distributions in a mouse were evaluated.Main results.The CT numbers from fastCAT agree within 11 HU with TOPAS except for materials at the centre of the phantom. Discrepancies for central inserts are caused by beam hardening issues. The root mean square deviation in the SPR for the best SECT (90 kV/Cu) and DECT (50 kV/Al-90 kV/Al) protocols are 3.7% and 1.0%, respectively. Dose distributions calculated for SECT and DECT datasets revealed range shifts <0.1 mm, gamma pass rates (3%/0.1 mm) greater than 99%, and no substantial dosimetric differences for all structures. The outcomes suggest that SECT is sufficient for proton treatment planning in animals.Significance.The framework is a useful tool for the development of an optimized experimental configuration without using animals and beam time.
Collapse
Affiliation(s)
- Justin Malimban
- Department of Radiation Oncology and Particle Therapy Research Center, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Felix Ludwig
- Department of Radiation Oncology and Particle Therapy Research Center, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Danny Lathouwers
- Department of Radiation Science and Technology, Faculty of Applied Sciences, Delft University of Technology, Delft, The Netherlands
| | - Marius Staring
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Radiation Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Frank Verhaegen
- Department of Radiation Oncology (MAASTRO), Research Institute for Oncology & Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Sytze Brandenburg
- Department of Radiation Oncology and Particle Therapy Research Center, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
21
|
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
|
22
|
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
|
23
|
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
|
24
|
van Marlen P, van de Water S, Slotman BJ, Dahele M, Verbakel W. Technical note: Dosimetry and FLASH potential of UHDR proton PBS for small lung tumors: Bragg-peak-based delivery versus transmission beam and IMPT. Med Phys 2024; 51:7580-7588. [PMID: 38795376 DOI: 10.1002/mp.17185] [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: 11/22/2023] [Revised: 04/19/2024] [Accepted: 05/04/2024] [Indexed: 05/27/2024] Open
Abstract
BACKGROUND High-energy transmission beams (TBs) are currently the main delivery method for proton pencil beam scanning ultrahigh dose-rate (UHDR) FLASH radiotherapy. TBs place the Bragg-peaks behind the target, outside the patient, making delivery practical and achievement of high dose-rates more likely. However, they lead to higher integral dose compared to conventional intensity-modulated proton therapy (IMPT), in which Bragg-peaks are placed within the tumor. It is hypothesized that, when energy changes are not required and high beam currents are possible, Bragg-peak-based beams can not only achieve more conformal dose distributions than TBs, but also have more FLASH-potential. PURPOSE This works aims to verify this hypothesis by taking three different Bragg-peak-based delivery techniques and comparing them with TB and IMPT-plans in terms of dosimetry and FLASH-potential for single-fraction lung stereotactic body radiotherapy (SBRT). METHODS For a peripherally located lung target of various sizes, five different proton plans were made using "matRad" and inhouse-developed algorithms for spot/energy-layer/beam reduction and minimum monitor unit maximization: (1) IMPT-plan, reference for dosimetry, (2) TB-plan, reference for FLASH-amount, (3) pristine Bragg-peak plan (non-depth-modulated Bragg-peaks), (4) Bragg-peak plan using generic ridge filter, and (5) Bragg-peak plan using 3D range-modulated ridge filter. RESULTS Bragg-peak-based plans are able to achieve sufficient plan quality and high dose-rates. IMPT-plans resulted in lowest OAR-dose and integral dose (also after a FLASH sparing-effect of 30%) compared to both TB-plans and Bragg-peak-based plans. Bragg-peak-based plans vary only slightly between themselves and generally achieve lower integral dose than TB-plans. However, TB-plans nearly always resulted in lower mean lung dose than Bragg-peak-based plans and due to a higher amount of FLASH-dose for TB-plans, this difference increased after including a FLASH sparing-effect. CONCLUSION This work indicates that there is no benefit in using Bragg-peak-based beams instead of TBs for peripherally located, UHDR stereotactic lung radiotherapy, if lung dose is the priority.
Collapse
Affiliation(s)
- Patricia van Marlen
- Department of Radiation Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Steven van de Water
- Department of Radiation Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Ben J Slotman
- Department of Radiation Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Max Dahele
- Department of Radiation Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | | |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Sørensen BS, Kanouta E, Ankjærgaard C, Kristensen L, Johansen JG, Sitarz MK, Andersen CE, Grau C, Poulsen P. Proton FLASH: Impact of Dose Rate and Split Dose on Acute Skin Toxicity in a Murine Model. Int J Radiat Oncol Biol Phys 2024; 120:265-275. [PMID: 38750904 DOI: 10.1016/j.ijrobp.2024.04.071] [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: 07/28/2023] [Revised: 04/04/2024] [Accepted: 04/25/2024] [Indexed: 06/01/2024]
Abstract
PURPOSE Preclinical studies have shown a preferential normal tissue sparing effect of FLASH radiation therapy with ultra-high dose rates. The aim of the present study was to use a murine model of acute skin toxicity to investigate the biologic effect of varying dose rates, time structure, and introducing pauses in the dose delivery. METHODS AND MATERIALS The right hind limbs of nonanaesthetized mice were irradiated in the entrance plateau of a pencil beam scanning proton beam with 39.3 Gy. Experiment 1 was with varying field dose rates (0.7-80 Gy/s) without repainting, experiment 2 was with varying field dose rates (0.37-80 Gy/s) with repainting, and in experiment 3, the dose was split into 2, 3, 4, or 6 identical deliveries with 2-minute pauses. In total, 320 mice were included, with 6 to 25 mice per group. The endpoints were skin toxicity of different levels up to 25 days after irradiation. RESULTS The dose rate50, which is the dose rate to induce a response in 50% of the animals, depended on the level of skin toxicity, with the higher toxicity levels displaying a FLASH effect at 0.7-2 Gy/s. Repainting resulted in higher toxicity for the same field dose rate. Splitting the dose into 2 deliveries reduced the FLASH effect, and for 3 or more deliveries, the FLASH effect was almost abolished for lower grades of toxicity. CONCLUSIONS The dose rate that induced a FLASH effect varied for different skin toxicity levels, which are characterized by a differing degree of sensitivity to radiation dosage. Conclusions on a threshold for the dose rate needed to obtain a FLASH effect can therefore be influenced by the dose sensitivity of the used endpoint. Splitting the total dose into more deliveries compromised the FLASH effect. This can have an impact for fractionation as well as for regions where 2 or more FLASH fields overlap within the same treatment session.
Collapse
Affiliation(s)
- Brita Singers Sørensen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Experimental Clinical Oncology, Aarhus University, Denmark; Department of Clinical Medicine, Health, AU; Department of Oncology, Aarhus University Hospital, Aarhus, Denmark.
| | - Eleni Kanouta
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Health, AU
| | | | - Line Kristensen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Experimental Clinical Oncology, Aarhus University, Denmark; Department of Clinical Medicine, Health, AU; Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jacob G Johansen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Health, AU
| | - Mateusz Krzysztof Sitarz
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Health, AU
| | | | - Cai Grau
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Health, AU
| | - Per Poulsen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Health, AU; Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
28
|
Præstegaard LH. Radiation safety of ultra-high dose rate electron accelerators for FLASH radiotherapy. Med Phys 2024; 51:6206-6219. [PMID: 38941539 DOI: 10.1002/mp.17245] [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: 02/26/2024] [Revised: 05/16/2024] [Accepted: 05/22/2024] [Indexed: 06/30/2024] Open
Abstract
BACKGROUND An ultra-high dose rate (UHDR) electron accelerator for FLASH radiotherapy (RT) produces very intense bremsstrahlung by the interaction of the electron beam with objects both inside and outside of the accelerator. The bremsstrahlung dose per pulse is typically 1-2 orders of magnitude larger than that of conventional RT x-ray treatment of the same energy, and for electron energies above 10 MeV, the bremsstrahlung produces substantially more induced radioactivity outside the accelerator than for conventional RT. Therefore, a thorough radiation safety assessment is mandatory prior to the operation of a UHDR electron accelerator. PURPOSE To evaluate the radiation safety of a prototype FLASH-enabled Varian TrueBeam accelerator and to develop a general framework for assessment of all key radiation safety properties of a UHDR electron accelerator for FLASH RT. METHODS Production of bremsstrahlung and induced radioactivity by a UHDR electron accelerator is modeled by various analytical methods. The analytical modeling is compared with National Institute of Standards and Technology (NIST) bremsstrahlung yield data as well as measurements of primary bremsstrahlung outside the bunker and induced radioactivity of irradiated thick targets for a FLASH-enabled 16 MeV Varian TrueBeam electron accelerator. In addition, the analytical modeling is complemented by measurements of secondary bremsstrahlung inside/outside the bunker and neutrons at the maze entrance. RESULTS Calculated bremsstrahlung yields deviate maximum 8.5% from NIST data, and all measurements of primary bremsstrahlung and induced radioactivity agree with calculations, validating the analytical tools. In addition, it is found that scattering foil bremsstrahlung dominates primary bremsstrahlung and the main source of secondary bremsstrahlung is the irradiated object outside the accelerator. It follows that primary and secondary bremsstrahlung outside the bunker can be calculated using the same simple formalism as that used for conventional RT. Measured primary bremsstrahlung tenth-value layers for concrete of the simple formalism are in good agreement with NCRP and IAEA data, while measured secondary bremsstrahlung tenth-value layers for concrete are considerably lower than NCRP and IAEA data. All calculations and measurements form a general framework for assessment of all key radiation safety properties of a UHDR electron accelerator. CONCLUSIONS The FLASH-enabled Varian TrueBeam accelerator is safe for normal operation (max. 99 pulses per irradiation) in a bunker designed for at least 15 MV conventional x-ray treatment unless the UHDR workload is much larger than the x-ray workload. A similar finding applies to other UHDR electron accelerators. However, during beam tuning, radiation survey, or other tests with extended irradiation time, the UHDR workload may become very large, necessitating the implementation of additional safety measures.
Collapse
|
29
|
Poulsen PR, Johansen JG, Sitarz MK, Kanouta E, Kristensen L, Grau C, Sørensen BS. Oxygen Enhancement Ratio-Weighted Dose Quantitatively Describes Acute Skin Toxicity Variations in Mice After Pencil Beam Scanning Proton FLASH Irradiation With Changing Doses and Time Structures. Int J Radiat Oncol Biol Phys 2024; 120:276-286. [PMID: 38462015 DOI: 10.1016/j.ijrobp.2024.02.050] [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: 07/30/2023] [Revised: 02/12/2024] [Accepted: 02/24/2024] [Indexed: 03/12/2024]
Abstract
PURPOSE The aim of this work was to investigate the ability of a biological oxygen enhancement ratio-weighted dose, DOER, to describe acute skin toxicity variations observed in mice after proton pencil beam scanning irradiations with changing doses and beam time structures. METHODS AND MATERIALS In five independent experiments, the right hind leg of a total of 621 CDF1 mice was irradiated previously in the entrance plateau of a pencil beam scanning proton beam. The incidence of acute skin toxicity (of level 1.5-2.0-2.5-3.0-3.5) was scored for 47 different mouse groups that mapped toxicity as function of dose for conventional and FLASH dose rate, toxicity as function of field dose rate with and without repainting, and toxicity when splitting the treatment into 1 to 6 identical deliveries separated by 2 minutes. DOER was calculated for all mouse groups using a simple oxygen kinetics model to describe oxygen depletion. The three independent model parameters (oxygen-depletion rate, oxygen-recovery rate, oxygen level without irradiation) were fitted to the experimental data. The ability of DOER to describe the toxicity variations across all experiments was investigated by comparing DOER-response curves across the five independent experiments. RESULTS After conversion from the independent variable tested in each experiment to DOER, all five experiments had similar MDDOER50 (DOER giving 50% toxicity incidence) with standard deviations of 0.45 - 1.6 Gy for the five toxicity levels. DOER could thus describe the observed toxicity variations across all experiments. CONCLUSIONS DOER described the varying FLASH-sparing effect observed for a wide range of conditions. Calculation of DOER for other irradiation conditions can quantitatively estimate the FLASH-sparing effect for arbitrary irradiations for the investigated murine model. With appropriate fitting parameters DOER also may be able to describe FLASH effect variations with dose and dose rate for other assays and endpoints.
Collapse
Affiliation(s)
- Per Rugaard Poulsen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Jacob Graversen Johansen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Mateusz Krzysztof Sitarz
- 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
| | - Line Kristensen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - 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 Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
30
|
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
|
31
|
Dai T, Sloop AM, Rahman MR, Sunnerberg JP, Clark MA, Young R, Adamczyk S, Voigts-Rhetz PV, Patane C, Turk M, Jarvis L, Pogue BW, Gladstone DJ, Bruza P, Zhang R. First Monte Carlo beam model for ultra-high dose rate radiotherapy with a compact electron LINAC. Med Phys 2024; 51:5109-5118. [PMID: 38493501 PMCID: PMC11316970 DOI: 10.1002/mp.17031] [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: 09/21/2023] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 03/19/2024] Open
Abstract
BACKGROUND FLASH radiotherapy based on ultra-high dose rate (UHDR) is actively being studied by the radiotherapy community. Dedicated UHDR electron devices are currently a mainstay for FLASH studies. PURPOSE To present the first Monte Carlo (MC) electron beam model for the UHDR capable Mobetron (FLASH-IQ) as a dose calculation and treatment planning platform for preclinical research and FLASH-radiotherapy (RT) clinical trials. METHODS The initial beamline geometry of the Mobetron was provided by the manufacturer, with the first-principal implementation realized in the Geant4-based GAMOS MC toolkit. The geometry and electron source characteristics, such as energy spectrum and beamline parameters, were tuned to match the central-axis percentage depth dose (PDD) and lateral profiles for the pristine beam measured during machine commissioning. The thickness of the small foil in secondary scatter affected the beam model dominantly and was fine tuned to achieve the best agreement with commissioning data. Validation of the MC beam modeling was performed by comparing the calculated PDDs and profiles with EBT-XD radiochromic film measurements for various combinations of applicators and inserts. RESULTS The nominal 9 MeV electron FLASH beams were best represented by a Gaussian energy spectrum with mean energy of 9.9 MeV and variance (σ) of 0.2 MeV. Good agreement between the MC beam model and commissioning data were demonstrated with maximal discrepancy < 3% for PDDs and profiles. Hundred percent gamma pass rate was achieved for all PDDs and profiles with the criteria of 2 mm/3%. With the criteria of 2 mm/2%, maximum, minimum and mean gamma pass rates were (100.0%, 93.8%, 98.7%) for PDDs and (100.0%, 96.7%, 99.4%) for profiles, respectively. CONCLUSIONS A validated MC beam model for the UHDR capable Mobetron is presented for the first time. The MC model can be utilized for direct dose calculation or to generate beam modeling input required for treatment planning systems for FLASH-RT planning. The beam model presented in this work should facilitate translational and clinical FLASH-RT for trials conducted on the Mobetron FLASH-IQ platform.
Collapse
Affiliation(s)
- Tianyuan Dai
- Thayer School of Engineering, Dartmouth College, Hanover NH 03755, USA
- 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, China
| | - Austin M. Sloop
- Thayer School of Engineering, Dartmouth College, Hanover NH 03755, USA
| | | | | | - Megan A. Clark
- Thayer School of Engineering, Dartmouth College, Hanover NH 03755, USA
| | - Ralph Young
- IntraOp Medical Corporation, Sunnyvale CA 94085, USA
| | | | | | - Chris Patane
- IntraOp Medical Corporation, Sunnyvale CA 94085, USA
| | - Michael Turk
- IntraOp Medical Corporation, Sunnyvale CA 94085, USA
| | - Lesley Jarvis
- Department of Medicine, Geisel School of Medicine, Dartmouth College Hanover NH 03755 USA
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756 USA
| | - Brian W. Pogue
- Thayer School of Engineering, Dartmouth College, Hanover NH 03755, USA
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756 USA
- Department of Medical Physics, Wisconsin Institutes for Medical Research, University of Wisconsin, Madison WI 53705 USA
| | - David J. Gladstone
- Thayer School of Engineering, Dartmouth College, Hanover NH 03755, USA
- Department of Medicine, Geisel School of Medicine, Dartmouth College Hanover NH 03755 USA
- Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756 USA
| | - Petr Bruza
- Thayer School of Engineering, Dartmouth College, Hanover NH 03755, USA
| | - Rongxiao Zhang
- Thayer School of Engineering, Dartmouth College, Hanover NH 03755, USA
- Department of Medicine, Geisel School of Medicine, Dartmouth College Hanover NH 03755 USA
- Department of Radiation Medicine, New York Medical College, Valhalla, NY 10595 USA
| |
Collapse
|
32
|
Loo BW, Verginadis II, Sørensen BS, Mascia AE, Perentesis JP, Koong AC, Schüler E, Rankin EB, Maxim PG, Limoli CL, Vozenin MC. Navigating the Critical Translational Questions for Implementing FLASH in the Clinic. Semin Radiat Oncol 2024; 34:351-364. [PMID: 38880544 DOI: 10.1016/j.semradonc.2024.04.008] [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] [Indexed: 06/18/2024]
Abstract
The "FLASH effect" is an increased therapeutic index, that is, reduced normal tissue toxicity for a given degree of anti-cancer efficacy, produced by ultra-rapid irradiation delivered on time scales orders of magnitude shorter than currently conventional in the clinic for the same doses. This phenomenon has been observed in numerous preclinical in vivo tumor and normal tissue models. While the underlying biological mechanism(s) remain to be elucidated, a path to clinical implementation of FLASH can be paved by addressing several critical translational questions. Technological questions pertinent to each beam type (eg, electron, proton, photon) also dictate the logical progression of experimentation required to move forward in safe and decisive clinical trials. Here we review the available preclinical data pertaining to these questions and how they may inform strategies for FLASH cancer therapy clinical trials.
Collapse
Affiliation(s)
- Billy W Loo
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA.
| | - Ioannis I Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Brita Singers Sørensen
- Danish Centre for Particle Therapy & Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Anthony E Mascia
- Division of Oncology, Cincinnati Children's Hospital and Departments of Pediatrics and Radiation Oncology, College of Medicine, University of Cincinnati, Cincinnati, OH
| | - John P Perentesis
- Division of Oncology, Cincinnati Children's Hospital and Departments of Pediatrics and Radiation Oncology, College of Medicine, University of Cincinnati, Cincinnati, OH
| | - Albert C Koong
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Emil Schüler
- Department of Radiation Physics, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Erinn B Rankin
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA; Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA
| | - Peter G Maxim
- Department of Radiation Oncology, University of California, Irvine School of Medicine, Irvine, CA
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine School of Medicine, Irvine, CA
| | - Marie-Catherine Vozenin
- Secteur Radio-Oncologie et Radiobiologie, Hôpitaux Universitaires de Genève, Geneva, Switzerland; LiRR - laboratory of innovation in radiobiology applied to radiotherapy, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Laboratory of Radiation Oncology, Radiation Oncology Service, Department of Oncology, CHUV Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
33
|
Kanouta E, Bruza P, Johansen JG, Kristensen L, Sørensen BS, Poulsen PR. Two-dimensional time-resolved scintillating sheet monitoring of proton pencil beam scanning FLASH mouse irradiations. Med Phys 2024; 51:5119-5129. [PMID: 38569159 DOI: 10.1002/mp.17049] [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: 09/19/2023] [Revised: 03/14/2024] [Accepted: 03/20/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Dosimetry in pre-clinical FLASH studies is essential for understanding the beam delivery conditions that trigger the FLASH effect. Resolving the spatial and temporal characteristics of proton pencil beam scanning (PBS) irradiations with ultra-high dose rates (UHDR) requires a detector with high spatial and temporal resolution. PURPOSE To implement a novel camera-based system for time-resolved two-dimensional (2D) monitoring and apply it in vivo during pre-clinical proton PBS mouse irradiations. METHODS Time-resolved 2D beam monitoring was performed with a scintillation imaging system consisting of a 1 mm thick transparent scintillating sheet, imaged by a CMOS camera. The sheet was placed in a water bath perpendicular to a horizontal PBS proton beam axis. The scintillation light was reflected through a system of mirrors and captured by the camera with 500 frames per second (fps) for UHDR and 4 fps for conventional dose rates. The raw images were background subtracted, geometrically transformed, flat field corrected, and spatially filtered. The system was used for 2D spot and field profile measurements and compared to radiochromic films. Furthermore, spot positions were measured for UHDR irradiations. The measured spot positions were compared to the planned positions and the relative instantaneous dose rate to equivalent fiber-coupled point scintillator measurements. For in vivo application, the scintillating sheet was placed 1 cm upstream the right hind leg of non-anaesthetized mice submerged in the water bath. The mouse leg and sheet were both placed in a 5 cm wide spread-out Bragg peak formed from the mono-energetic proton beam by a 2D range modulator. The mouse leg position within the field was identified for both conventional and FLASH irradiations. For the conventional irradiations, the mouse foot position was tracked throughout the beam delivery, which took place through repainting. For FLASH irradiations, the delivered spot positions and relative instantaneous dose rate were measured. RESULTS The pixel size was 0.1 mm for all measurements. The spot and field profiles measured with the scintillating sheet agreed with radiochromic films within 0.4 mm. The standard deviation between measured and planned spot positions was 0.26 mm and 0.35 mm in the horizontal and vertical direction, respectively. The measured relative instantaneous dose rate showed a linear relation with the fiber-coupled scintillator measurements. For in vivo use, the leg position within the field varied between mice, and leg movement up to 3 mm was detected during the prolonged conventional irradiations. CONCLUSIONS The scintillation imaging system allowed for monitoring of UHDR proton PBS delivery in vivo with 0.1 mm pixel size and 2 ms temporal resolution. The feasibility of instantaneous dose rate measurements was demonstrated, and the system was used for validation of the mouse leg position within the field.
Collapse
Affiliation(s)
- Eleni Kanouta
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Petr Bruza
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Jacob Graversen Johansen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Line Kristensen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Brita Singers Sørensen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Per Rugaard Poulsen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
34
|
Yan O, Wang S, Wang Q, Wang X. FLASH Radiotherapy: Mechanisms of Biological Effects and the Therapeutic Potential in Cancer. Biomolecules 2024; 14:754. [PMID: 39062469 PMCID: PMC11275005 DOI: 10.3390/biom14070754] [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: 05/21/2024] [Revised: 06/11/2024] [Accepted: 06/15/2024] [Indexed: 07/28/2024] Open
Abstract
Radiotherapy is an important treatment for many unresectable advanced malignant tumors, and radiotherapy-associated inflammatory reactions to radiation and other toxic side effects are significant reasons which reduce the quality of life and survival of patients. FLASH-radiotherapy (FLASH-RT), a prominent topic in recent radiation therapy research, is an ultra-high dose rate treatment known for significantly reducing therapy time while effectively targeting tumors. This approach minimizes radiation side effects on at-risk organs and maximally protects surrounding healthy tissues. Despite decades of preclinical exploration and some notable achievements, the mechanisms behind FLASH effects remain debated. Standardization is still required for the type of FLASH-RT rays and dose patterns. This review addresses the current state of FLASH-RT research, summarizing the biological mechanisms behind the FLASH effect. Additionally, it examines the impact of FLASH-RT on immune cells, cytokines, and the tumor immune microenvironment. Lastly, this review will discuss beam characteristics, potential clinical applications, and the relevance and applicability of FLASH-RT in treating advanced cancers.
Collapse
Affiliation(s)
| | | | | | - Xin Wang
- Division of Abdominal Tumor Multimodality Treatment, Department of Radiation Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu 610041, China; (O.Y.); (S.W.); (Q.W.)
| |
Collapse
|
35
|
Lövgren N, Fagerström Kristensen I, Petersson K. Feasibility and constraints of Bragg peak FLASH proton therapy treatment planning. Front Oncol 2024; 14:1369065. [PMID: 38737902 PMCID: PMC11082391 DOI: 10.3389/fonc.2024.1369065] [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: 01/11/2024] [Accepted: 04/02/2024] [Indexed: 05/14/2024] Open
Abstract
Introduction FLASH proton therapy (FLASH-PT) requires ultra-high dose rate (≥ 40 Gy/s) protons to be delivered in a short timescale whilst conforming to a patient-specific target. This study investigates the feasibility and constraints of Bragg peak FLASH-PT treatment planning, and compares the in silico results produced to plans for intensity modulated proton therapy (IMPT). Materials and method Bragg peak FLASH-PT and IMPT treatment plans were generated for bone (n=3), brain (n=3), and lung (n=4) targets using the MIROpt research treatment planning system and the Conformal FLASH library developed by Applications SA from the open-source version of UCLouvain. FLASH-PT beams were simulated using monoenergetic spot-scanned protons traversing through a conformal energy modulator, a range shifter, and an aperture. A dose rate constraint of ≥ 40 Gy/s was included in each FLASH-PT plan optimisation. Results Space limitations in the FLASH-PT adapted beam nozzle imposed a maximum target width constraint, excluding 4 cases from the study. FLASH-PT plans did not satisfy the imposed target dose constraints (D95% ≥ 95% and D2%≤ 105%) but achieved clinically acceptable doses to organs at risk (OARs). IMPT plans adhered to all target and OAR dose constraints. FLASH-PT plans showed a reduction in both target homogeneity (p < 0.001) and dose conformity (non-significant) compared to IMPT. Conclusion Without accounting for a sparing effect, IMPT plans were superior in target coverage, dose conformity, target homogeneity, and OAR sparing compared to FLASH-PT. Further research is warranted in treatment planning optimisation and beam delivery for clinical implementation of Bragg peak FLASH-PT.
Collapse
Affiliation(s)
- Nathalie Lövgren
- Department of Oncology, Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Ingrid Fagerström Kristensen
- Clinical Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Department of Haematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Kristoffer Petersson
- Department of Oncology, Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
- Department of Haematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
36
|
McGarrigle JM, Long KR, Prezado Y. The FLASH effect-an evaluation of preclinical studies of ultra-high dose rate radiotherapy. Front Oncol 2024; 14:1340190. [PMID: 38711846 PMCID: PMC11071325 DOI: 10.3389/fonc.2024.1340190] [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: 11/17/2023] [Accepted: 03/20/2024] [Indexed: 05/08/2024] Open
Abstract
FLASH radiotherapy (FLASH-RT) is a novel radiotherapy approach based on the use of ultra-high dose radiation to treat malignant cells. Although tumours can be reduced or eradicated using radiotherapy, toxicities induced by radiation can compromise healthy tissues. The FLASH effect is the observation that treatment delivered at an ultra-high dose rate is able to reduce adverse toxicities present at conventional dose rates. While this novel technique may provide a turning point for clinical practice, the exact mechanisms underlying the causes or influences of the FLASH effect are not fully understood. The study presented here uses data collected from 41 experimental investigations (published before March 2024) of the FLASH effect. Searchable databases were constructed to contain the outcomes of the various experiments in addition to values of beam parameters that may have a bearing on the FLASH effect. An in-depth review of the impact of the key beam parameters on the results of the experiments was carried out. Correlations between parameter values and experimental outcomes were studied. Pulse Dose Rate had positive correlations with almost all end points, suggesting viability of FLASH-RT as a new modality of radiotherapy. The collective results of this systematic review study suggest that beam parameter qualities from both FLASH and conventional radiotherapy can be valuable for tissue sparing and effective tumour treatment.
Collapse
Affiliation(s)
| | - Kenneth Richard Long
- Department of Physics, Imperial College London, London, United Kingdom
- Science and Technology Facilities Council (STFC), Rutherford Appleton Laboratory, Oxford, United Kingdom
| | - Yolanda Prezado
- Institut Curie, Universite Paris-Saclay, Centre national de la recherche scientifique (CNRS) UMR3347, Inserm U1021, Signalisation radiobiologie et cancer, Orsay, France
- Universite Paris-Saclay, Centre national de la recherche scientifique (CNRS) UMR3347, Inserm U1021, Signalisation radiobiologie et cancer, Orsay, France
| |
Collapse
|
37
|
Tang R, Yin J, Liu Y, Xue J. FLASH radiotherapy: A new milestone in the field of cancer radiotherapy. Cancer Lett 2024; 587:216651. [PMID: 38342233 DOI: 10.1016/j.canlet.2024.216651] [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: 08/15/2023] [Revised: 11/03/2023] [Accepted: 01/13/2024] [Indexed: 02/13/2024]
Abstract
Radiotherapy plays a pivotal role in the control and eradication of tumors, but it can also induce radiation injury to surrounding normal tissues while targeting tumor cells. In recent years, FLASH-Radiotherapy (FLASH-RT) has emerged as a cutting-edge research focus in the field of radiation therapy. By delivering high radiation doses to the treatment target in an ultra-short time, FLASH-RT produces the FLASH effect, which reduces the toxicity to normal tissues while achieving comparable tumor control efficacy to conventional radiotherapy. This review provides a brief overview of the development history of FLASH-RT and its impact on tumor control. Additionally, it focuses on introducing the protective effects and molecular mechanisms of this technology on various normal tissues, as well as exploring its synergistic effects when combined with other tumor therapies. Importantly, this review discusses the challenges faced in translating FLASH-RT into clinical practice and outlines its promising future applications.
Collapse
Affiliation(s)
- Rui Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan, China; Division of Thoracic Tumor Multimodality Treatment, Cancer Center, The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jianqiong Yin
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yuanxin Liu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jianxin Xue
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Disaster Medical Center, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
38
|
Levin DS, Friedman PS, Ferretti C, Ristow N, Tecchio M, Litzenberg DW, Bashkirov V, Schulte R. A prototype scintillator real-time beam monitor for ultra-high dose rate radiotherapy. Med Phys 2024; 51:2905-2923. [PMID: 38456622 PMCID: PMC11992679 DOI: 10.1002/mp.17018] [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: 05/24/2023] [Revised: 02/24/2024] [Accepted: 02/26/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND FLASH Radiotherapy (RT) is an emergent cancer RT modality where an entire therapeutic dose is delivered at more than 1000 times higher dose rate than conventional RT. For clinical trials to be conducted safely, a precise and fast beam monitor that can generate out-of-tolerance beam interrupts is required. This paper describes the overall concept and provides results from a prototype ultra-fast, scintillator-based beam monitor for both proton and electron beam FLASH applications. PURPOSE A FLASH Beam Scintillator Monitor (FBSM) is being developed that employs a novel proprietary scintillator material. The FBSM has capabilities that conventional RT detector technologies are unable to simultaneously provide: (1) large area coverage; (2) a low mass profile; (3) a linear response over a broad dynamic range; (4) radiation hardness; (5) real-time analysis to provide an IEC-compliant fast beam-interrupt signal based on true two-dimensional beam imaging, radiation dosimetry and excellent spatial resolution. METHODS The FBSM uses a proprietary low mass, less than 0.5 mm water equivalent, non-hygroscopic, radiation tolerant scintillator material (designated HM: hybrid material) that is viewed by high frame rate CMOS cameras. Folded optics using mirrors enable a thin monitor profile of ∼10 cm. A field programmable gate array (FPGA) data acquisition system generates real-time analysis on a time scale appropriate to the FLASH RT beam modality: 100-1000 Hz for pulsed electrons and 10-20 kHz for quasi-continuous scanning proton pencil beams. An ion beam monitor served as the initial development platform for this work and was tested in low energy heavy-ion beams (86Kr+26 and protons). A prototype FBSM was fabricated and then tested in various radiation beams that included FLASH level dose per pulse electron beams, and a hospital RT clinic with electron beams. RESULTS Results presented in this report include image quality, response linearity, radiation hardness, spatial resolution, and real-time data processing. The HM scintillator was found to be highly radiation damage resistant. It exhibited a small 0.025%/kGy signal decrease from a 216 kGy cumulative dose resulting from continuous exposure for 15 min at a FLASH compatible dose rate of 237 Gy/s. Measurements of the signal amplitude versus beam fluence demonstrate linear response of the FBSM at FLASH compatible dose rates of >40 Gy/s. Comparison with commercial Gafchromic film indicates that the FBSM produces a high resolution 2D beam image and can reproduce a nearly identical beam profile, including primary beam tails. The spatial resolution was measured at 35-40 µm. Tests of the firmware beta version show successful operation at 20 000 Hz frame rate or 50 µs/frame, where the real-time analysis of the beam parameters is achieved in less than 1 µs. CONCLUSIONS The FBSM is designed to provide real-time beam profile monitoring over a large active area without significantly degrading the beam quality. A prototype device has been staged in particle beams at currents of single particles up to FLASH level dose rates, using both continuous ion beams and pulsed electron beams. Using a novel scintillator, beam profiling has been demonstrated for currents extending from single particles to 10 nA currents. Radiation damage is minimal and even under FLASH conditions would require ≥50 kGy of accumulated exposure in a single spot to result in a 1% decrease in signal output. Beam imaging is comparable to radiochromic films, and provides immediate images without hours of processing. Real-time data processing, taking less than 50 µs (combined data transfer and analysis times), has been implemented in firmware for 20 kHz frame rates for continuous proton beams.
Collapse
Affiliation(s)
- Daniel S. Levin
- Department of Physics, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Claudio Ferretti
- Department of Physics, University of Michigan, Ann Arbor, Michigan, USA
| | - Nicholas Ristow
- Department of Physics, University of Michigan, Ann Arbor, Michigan, USA
| | - Monica Tecchio
- Department of Physics, University of Michigan, Ann Arbor, Michigan, USA
| | - Dale W. Litzenberg
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Vladimir Bashkirov
- Division of Biomedical Engineering Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Reinhard Schulte
- Division of Biomedical Engineering Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| |
Collapse
|
39
|
Cengel KA, Kim MM, Diffenderfer ES, Busch TM. FLASH Radiotherapy: What Can FLASH's Ultra High Dose Rate Offer to the Treatment of Patients With Sarcoma? Semin Radiat Oncol 2024; 34:218-228. [PMID: 38508786 DOI: 10.1016/j.semradonc.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
FLASH is an emerging treatment paradigm in radiotherapy (RT) that utilizes ultra-high dose rates (UHDR; >40 Gy)/s) of radiation delivery. Developing advances in technology support the delivery of UHDR using electron and proton systems, as well as some ion beam units (eg, carbon ions), while methods to achieve UHDR with photons are under investigation. The major advantage of FLASH RT is its ability to increase the therapeutic index for RT by shifting the dose response curve for normal tissue toxicity to higher doses. Numerous preclinical studies have been conducted to date on FLASH RT for murine sarcomas, alongside the investigation of its effects on relevant normal tissues of skin, muscle, and bone. The tumor control achieved by FLASH RT of sarcoma models is indistinguishable from that attained by treatment with standard RT to the same total dose. FLASH's high dose rates are able to mitigate the severity or incidence of RT side effects on normal tissues as evaluated by endpoints ranging from functional sparing to histological damage. Large animal studies and clinical trials of canine patients show evidence of skin sparing by FLASH vs. standard RT, but also caution against delivery of high single doses with FLASH that exceed those safely applied with standard RT. Also, a human clinical trial has shown that FLASH RT can be delivered safely to bone metastasis. Thus, data to date support continued investigations of clinical translation of FLASH RT for the treatment of patients with sarcoma. Toward this purpose, hypofractionated irradiation schemes are being investigated for FLASH effects on sarcoma and relevant normal tissues.
Collapse
Affiliation(s)
- Keith A Cengel
- 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
| | - Eric S Diffenderfer
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Theresa M Busch
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
40
|
Yin L, Masumi U, Ota K, Sforza DM, Miles D, Rezaee M, Wong JW, Jia X, Li H. Feasibility of Synchrotron-Based Ultra-High Dose Rate (UHDR) Proton Irradiation with Pencil Beam Scanning for FLASH Research. Cancers (Basel) 2024; 16:221. [PMID: 38201648 PMCID: PMC10778151 DOI: 10.3390/cancers16010221] [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: 11/27/2023] [Revised: 12/12/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND This study aims to present the feasibility of developing a synchrotron-based proton ultra-high dose rate (UHDR) pencil beam scanning (PBS) system. METHODS The RF extraction power in the synchrotron system was increased to generate 142.4 MeV pulsed proton beams for UHDR irradiation at ~100 nA beam current. The charge per spill was measured using a Faraday cup. The spill length and microscopic time structure of each spill was measured with a 2D strip transmission ion chamber. The measured UHDR beam fluence was used to derive the spot dwell time for pencil beam scanning. Absolute dose distributions at various depths and spot spacings were measured using Gafchromic films in a solid-water phantom. RESULTS For proton UHDR beams at 142.4 MeV, the maximum charge per spill is 4.96 ± 0.10 nC with a maximum spill length of 50 ms. This translates to an average beam current of approximately 100 nA during each spill. Using a 2 × 2 spot delivery pattern, the delivered dose per spill at 5 cm and 13.5 cm depth is 36.3 Gy (726.3 Gy/s) and 56.2 Gy (1124.0 Gy/s), respectively. CONCLUSIONS The synchrotron-based proton therapy system has the capability to deliver pulsed proton UHDR PBS beams. The maximum deliverable dose and field size per pulse are limited by the spill length and extraction charge.
Collapse
Affiliation(s)
- Lingshu Yin
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (D.M.S.); (D.M.); (M.R.); (J.W.W.); (X.J.); (H.L.)
| | - Umezawa Masumi
- Hitachi, Ltd., Research and Development Group, Center for Technology Innovation–Energy, 7-2-1, Omika-chou, Hitachi-shi 319-1292, Ibaraki-ken, Japan;
| | - Kan Ota
- Pyramid Technical Consultants, Inc., Boston, MA 02452, USA;
| | - Daniel M. Sforza
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (D.M.S.); (D.M.); (M.R.); (J.W.W.); (X.J.); (H.L.)
| | - Devin Miles
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (D.M.S.); (D.M.); (M.R.); (J.W.W.); (X.J.); (H.L.)
| | - Mohammad Rezaee
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (D.M.S.); (D.M.); (M.R.); (J.W.W.); (X.J.); (H.L.)
| | - John W. Wong
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (D.M.S.); (D.M.); (M.R.); (J.W.W.); (X.J.); (H.L.)
| | - Xun Jia
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (D.M.S.); (D.M.); (M.R.); (J.W.W.); (X.J.); (H.L.)
| | - Heng Li
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (D.M.S.); (D.M.); (M.R.); (J.W.W.); (X.J.); (H.L.)
| |
Collapse
|
41
|
Wu X, Luo H, Wang Q, Du T, Chen Y, Tan M, Liu R, Liu Z, Sun S, Yang K, Tian J, Zhang Q. Examining the Occurrence of the FLASH Effect in Animal Models: A Systematic Review and Meta-Analysis of Ultra-High Dose Rate Proton or Carbon Ion Irradiation. Technol Cancer Res Treat 2024; 23:15330338241289990. [PMID: 39512217 PMCID: PMC11544673 DOI: 10.1177/15330338241289990] [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: 07/06/2024] [Revised: 08/19/2024] [Accepted: 09/02/2024] [Indexed: 11/15/2024] Open
Abstract
Purpose: This systematic review and meta-analysis sought to assess whether ultra-high dose rate (UHDR) ion irradiations can induce the FLASH effect in animal models. Methods: A comprehensive search of the Web of Science, PubMed, and EMBASE databases was conducted from inception until March 20, 2023, to identify studies involving irradiated animals subjected to proton or carbon ion beams at varying dose rates. The research content should include various indicators that can reflect the effect and safety of radiation, such as survival, normal tissue toxicity, inflammatory response, tumor volume, etc Results: Compared to conventional dose rate (CONV) ion irradiations, UHDR ion irradiations can significantly improve mouse survival (HR 0.48, 95% CI 0.29 to 0.78, I2 = 0%) and maintain comparable tumor control. There was no significant impact of different dose rates on the survival of zebrafish embryos (SMD 0.11, 95% CI -0.31 to 0.53, I2 = 85%). Subgroup analysis showed that radiation dose was an important factor affecting the survival of zebrafish embryos. Achieving normal tissue sparing may require higher radiation dose under UHD.In mouse and zebrafish embryo models, normal tissue sparing did not always occur after UHDR ion irradiations. In addition, only a limited number of cytokines (CXCL1, IL-6, GM-CSF, G-CSF, HMGB1, and TGF-β) and immune cells (microglia and myeloid cells) showed differences at different dose rates. Conclusions: UHDR ion irradiation can achieve FLASH effect, but the reproducibility of normal tissue sparing remains a challenge. Compared to CONV irradiation, UHDR ion irradiations demonstrated equivalent or even superior tumor control.
Collapse
Affiliation(s)
- Xun Wu
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Chengguan District, Lanzhou 730000, People's Republic of China
- The First School of Clinical Medicine, Lanzhou University, No.1 Donggang West Road, Chengguan District, Lanzhou 730000, People's Republic of China
| | - Hongtao Luo
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Chengguan District, Lanzhou 730000, People's Republic of China
| | - Qian Wang
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Chengguan District, Lanzhou 730000, People's Republic of China
- The First School of Clinical Medicine, Lanzhou University, No.1 Donggang West Road, Chengguan District, Lanzhou 730000, People's Republic of China
| | - Tianqi Du
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Chengguan District, Lanzhou 730000, People's Republic of China
- The First School of Clinical Medicine, Lanzhou University, No.1 Donggang West Road, Chengguan District, Lanzhou 730000, People's Republic of China
| | - Yanliang Chen
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Chengguan District, Lanzhou 730000, People's Republic of China
- The First School of Clinical Medicine, Lanzhou University, No.1 Donggang West Road, Chengguan District, Lanzhou 730000, People's Republic of China
| | - Mingyu Tan
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Chengguan District, Lanzhou 730000, People's Republic of China
- The First School of Clinical Medicine, Lanzhou University, No.1 Donggang West Road, Chengguan District, Lanzhou 730000, People's Republic of China
| | - Ruifeng Liu
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Chengguan District, Lanzhou 730000, People's Republic of China
| | - Zhiqiang Liu
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Chengguan District, Lanzhou 730000, People's Republic of China
| | - Shilong Sun
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Chengguan District, Lanzhou 730000, People's Republic of China
| | - Kehu Yang
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, 222 Tianshui South Road, Chengguan District, Lanzhou 730000, People's Republic of China
| | - Jinhui Tian
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, 222 Tianshui South Road, Chengguan District, Lanzhou 730000, People's Republic of China
| | - Qiuning Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Chengguan District, Lanzhou 730000, People's Republic of China
- Graduate School, University of Chinese Academy of Sciences, 19A Yuquan Road, Shijingshan District, Beijing 100000, People's Republic of China
| |
Collapse
|
42
|
Mascia A, McCauley S, Speth J, Nunez SA, Boivin G, Vilalta M, Sharma RA, Perentesis JP, Sertorio M. Impact of Multiple Beams on the FLASH Effect in Soft Tissue and Skin in Mice. Int J Radiat Oncol Biol Phys 2024; 118:253-261. [PMID: 37541394 DOI: 10.1016/j.ijrobp.2023.07.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 06/19/2023] [Accepted: 07/14/2023] [Indexed: 08/06/2023]
Abstract
PURPOSE FLASH proton pencil beam scanning (p-PBS) showed a reduction in mouse skin toxicity and fibrosis when delivered as a single, uninterrupted, high-dose fraction. Clinical p-PBS treatment usually requires multiple beams to achieve good conformality, and these beams are separated by minutes to allow patient and equipment repositioning. We evaluate the impact of multibeam versus single-beam proton radiation on the FLASH sparing effect on skin toxicity. METHODS AND MATERIALS The right hind leg of 10-week-old female C57Bl/6j mice was irradiated using a Varian ProBeam proton beam scanning gantry system at conventional (1 Gy/s) or FLASH (100 Gy/s) average field dose rate. We scored the skin toxicity after different doses for 7 weeks. The treatment was delivered as 1, 2, or 3 equal beams with an interruption of 2 minutes. For each beam delivery, the equipment remained in the same position so that there was a full overlap of beams administered. RESULTS Single-beam delivery confirmed a benefit for p-PBS FLASH in this model at 30, 35, and 40 Gy. At 30 and 35 Gy, a single beam interruption of 2 minutes (2 × 15 Gy or 2 × 17.5 Gy) reduced the FLASH sparing effect, which remained significant (P < .001). However, 2 interruptions (3 × 10 Gy or 3 × 11.6 Gy) abrogated the normal tissue sparing effect. CONCLUSIONS Our results indicate that the FLASH sparing effect in areas of beam overlap can be compromised by interruptions in delivery time. Time gap between overlapping beams and spatial arrangement of the delivered beams are important parameters for FLASH studies. The effect of multibeam needs to be studied on different organs of interest.
Collapse
Affiliation(s)
- Anthony Mascia
- Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnati, Ohio; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Shelby McCauley
- Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Joseph Speth
- Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnati, Ohio; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Stefanno Alarcon Nunez
- Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnati, Ohio; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Gael Boivin
- Varian, a Siemens Healthineers Company, Palo Alto, California
| | - Marta Vilalta
- Varian, a Siemens Healthineers Company, Palo Alto, California
| | - Ricky A Sharma
- Varian, a Siemens Healthineers Company, Palo Alto, California
| | | | - Mathieu Sertorio
- Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnati, Ohio; Department of Radiation Oncology, University of Cincinnati Cancer Center, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| |
Collapse
|
43
|
Böhlen TT, Germond JF, Petersson K, Ozsahin EM, Herrera FG, Bailat C, Bochud F, Bourhis J, Moeckli R, Adrian G. Effect of Conventional and Ultrahigh Dose Rate FLASH Irradiations on Preclinical Tumor Models: A Systematic Analysis. Int J Radiat Oncol Biol Phys 2023; 117:1007-1017. [PMID: 37276928 DOI: 10.1016/j.ijrobp.2023.05.045] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 04/19/2023] [Accepted: 05/26/2023] [Indexed: 06/07/2023]
Abstract
PURPOSE Compared with conventional dose rate irradiation (CONV), ultrahigh dose rate irradiation (UHDR) has shown superior normal tissue sparing. However, a clinically relevant widening of the therapeutic window by UHDR, termed "FLASH effect," also depends on the tumor toxicity obtained by UHDR. Based on a combined analysis of published literature, the current study examined the hypothesis of tumor isoefficacy for UHDR versus CONV and aimed to identify potential knowledge gaps to inspire future in vivo studies. METHODS AND MATERIALS A systematic literature search identified publications assessing in vivo tumor responses comparing UHDR and CONV. Qualitative and quantitative analyses were performed, including combined analyses of tumor growth and survival data. RESULTS We identified 66 data sets from 15 publications that compared UHDR and CONV for tumor efficacy. The median number of animals per group was 9 (range 3-15) and the median follow-up period was 30.5 days (range 11-230) after the first irradiation. Tumor growth assays were the predominant model used. Combined statistical analyses of tumor growth and survival data are consistent with UHDR isoefficacy compared with CONV. Only 1 study determined tumor-controlling dose (TCD50) and reported statistically nonsignificant differences. CONCLUSIONS The combined quantitative analyses of tumor responses support the assumption of UHDR isoefficacy compared with CONV. However, the comparisons are primarily based on heterogeneous tumor growth assays with limited numbers of animals and short follow-up, and most studies do not assess long-term tumor control probability. Therefore, the assays may be insensitive in resolving smaller response differences, such as responses of radioresistant tumor subclones. Hence, tumor cure experiments, including additional TCD50 experiments, are needed to confirm the assumption of isoeffectiveness in curative settings.
Collapse
Affiliation(s)
- Till Tobias Böhlen
- Institute of Radiation Physics, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Jean-François Germond
- Institute of Radiation Physics, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Kristoffer Petersson
- Department of Hematology, Oncology, and Radiation Physics, Skåne University Hospital, Lund, Sweden; MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Esat Mahmut Ozsahin
- Department of Radiation Oncology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Fernanda G Herrera
- Department of Radiation Oncology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Claude Bailat
- Institute of Radiation Physics, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - François Bochud
- Institute of Radiation Physics, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Jean Bourhis
- Department of Radiation Oncology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Raphaël Moeckli
- Institute of Radiation Physics, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland.
| | - Gabriel Adrian
- Department of Hematology, Oncology, and Radiation Physics, Skåne University Hospital, Lund, Sweden; Division of Oncology and Pathology, Department of Clinical Sciences, Skåne University Hospital, Lund University, Lund, Sweden
| |
Collapse
|
44
|
Konradsson E, Szecsenyi RE, Adrian G, Coskun M, Børresen B, Arendt ML, Erhart K, Bäck SÅ, Petersson K, Ceberg C. Evaluation of intensity-modulated electron FLASH radiotherapy in a clinical setting using veterinary cases. Med Phys 2023; 50:6569-6579. [PMID: 37696040 DOI: 10.1002/mp.16737] [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: 04/24/2023] [Revised: 08/08/2023] [Accepted: 08/29/2023] [Indexed: 09/13/2023] Open
Abstract
PURPOSE The increased normal tissue tolerance for FLASH radiotherapy (FLASH-RT), as compared to conventional radiotherapy, was first observed in ultra-high dose rate electron beams. Initial clinical trials in companion animals have revealed a high risk of developing osteoradionecrosis following high-dose single-fraction electron FLASH-RT, which may be related to inhomogeneities in the dose distribution. In the current study, we aim to evaluate the possibilities of intensity-modulated electron FLASH-RT in a clinical setting to ensure a homogeneous dose distribution in future veterinary and human clinical trials. METHODS Our beam model in the treatment planning system electronRT (.decimal, LLC, Sanford, FL, USA) was based on a 10-MeV electron beam from a clinical linear accelerator used to treat veterinary patients with FLASH-RT in a clinical setting. In electronRT, the beam can be intensity-modulated using tungsten island blocks in the electron block cutout, and range-modulated using a customized bolus with variable thickness. Modulations were first validated in a heterogeneous phantom by comparing measured and calculated dose distributions. To evaluate the impact of intensity modulation in superficial single-fraction FLASH-RT, a treatment planning study was conducted, including eight canine cancer patient cases with simulated tumors in the head-and-neck region. For each case, treatment plans with and without intensity modulation were created for a uniform bolus and a range-modulating bolus. Treatment plans were evaluated using a target dose homogeneity index (HI), a conformity index (CI), the near-maximum dose outside the target (D 2 % , Body - PTV ${D_{2{\mathrm{\% }},{\mathrm{\ Body}} - {\mathrm{PTV}}}}$ ), and the near-minimum dose to the target (D 98 % ${D_{98\% }}$ ). RESULTS By adding intensity modulation to plans with a uniform bolus, the HI could be improved (p = 0.017). The combination of a range-modulating bolus and intensity modulation provided a further significant improvement of the HI as compared to using intensity modulation in combination with a uniform bolus (p = 0.036). The range-modulating bolus also improved the CI compared to using a uniform bolus, both with an open beam (p = 0.046) and with intensity modulation (p = 0.018), as well as increased theD 98 % ${D_{98\% }}$ (p = 0.036 with open beam and p = 0.05 with intensity modulation) and reduced the medianD 2 % , Body - PTV ${D_{2\% ,{\mathrm{\ Body}} - {\mathrm{PTV}}}}$ (not significant). CONCLUSIONS By using intensity-modulated electron FLASH-RT in combination with range-modulating bolus, the target dose homogeneity and conformity in canine patients with simulated tumors in complex areas in the head-and-neck region could be improved. By utilizing this technique, we hope to decrease the dose outside the target volume and avoid hot spots in future clinical electron FLASH-RT studies, thereby reducing the risk of radiation-induced toxicity.
Collapse
Affiliation(s)
- Elise Konradsson
- Medical Radiation Physics, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Rebecka Ericsson Szecsenyi
- Medical Radiation Physics, Department of Clinical Sciences, Lund University, Lund, Sweden
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Gabriel Adrian
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Mizgin Coskun
- Medical Radiation Physics, Department of Clinical Sciences, Lund University, Lund, Sweden
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Betina Børresen
- Department of Veterinary Clinical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Maja Louise Arendt
- Department of Veterinary Clinical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Sven Åj Bäck
- Medical Radiation Physics, Department of Clinical Sciences, Lund University, Lund, Sweden
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| | - Kristoffer Petersson
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
- Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Crister Ceberg
- Medical Radiation Physics, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
45
|
Zou W, Zhang R, Schüler E, Taylor PA, Mascia AE, Diffenderfer ES, Zhao T, Ayan AS, Sharma M, Yu SJ, Lu W, Bosch WR, Tsien C, Surucu M, Pollard-Larkin JM, Schuemann J, Moros EG, Bazalova-Carter M, Gladstone DJ, Li H, Simone CB, Petersson K, Kry SF, Maity A, Loo BW, Dong L, Maxim PG, Xiao Y, Buchsbaum JC. Framework for Quality Assurance of Ultrahigh Dose Rate Clinical Trials Investigating FLASH Effects and Current Technology Gaps. Int J Radiat Oncol Biol Phys 2023; 116:1202-1217. [PMID: 37121362 PMCID: PMC10526970 DOI: 10.1016/j.ijrobp.2023.04.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/28/2023] [Accepted: 04/17/2023] [Indexed: 05/02/2023]
Abstract
FLASH radiation therapy (FLASH-RT), delivered with ultrahigh dose rate (UHDR), may allow patients to be treated with less normal tissue toxicity for a given tumor dose compared with currently used conventional dose rate. Clinical trials are being carried out and are needed to test whether this improved therapeutic ratio can be achieved clinically. During the clinical trials, quality assurance and credentialing of equipment and participating sites, particularly pertaining to UHDR-specific aspects, will be crucial for the validity of the outcomes of such trials. This report represents an initial framework proposed by the NRG Oncology Center for Innovation in Radiation Oncology FLASH working group on quality assurance of potential UHDR clinical trials and reviews current technology gaps to overcome. An important but separate consideration is the appropriate design of trials to most effectively answer clinical and scientific questions about FLASH. This paper begins with an overview of UHDR RT delivery methods. UHDR beam delivery parameters are then covered, with a focus on electron and proton modalities. The definition and control of safe UHDR beam delivery and current and needed dosimetry technologies are reviewed and discussed. System and site credentialing for large, multi-institution trials are reviewed. Quality assurance is then discussed, and new requirements are presented for treatment system standard analysis, patient positioning, and treatment planning. The tables and figures in this paper are meant to serve as reference points as we move toward FLASH-RT clinical trial performance. Some major questions regarding FLASH-RT are discussed, and next steps in this field are proposed. FLASH-RT has potential but is associated with significant risks and complexities. We need to redefine optimization to focus not only on the dose but also on the dose rate in a manner that is robust and understandable and that can be prescribed, validated, and confirmed in real time. Robust patient safety systems and access to treatment data will be critical as FLASH-RT moves into the clinical trials.
Collapse
Affiliation(s)
- Wei Zou
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA.
| | - Rongxiao Zhang
- Department of Radiation Oncology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Emil Schüler
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paige A Taylor
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Eric S Diffenderfer
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Tianyu Zhao
- Department of Radiation Oncology, Washington University, St. Louis, MO, USA
| | - Ahmet S Ayan
- Department of Radiation Oncology, Ohio State University, Columbus, OH, USA
| | - Manju Sharma
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Shu-Jung Yu
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Weiguo Lu
- Department of Radiation Oncology, University of Texas Southwestern, Dallas, TX, USA
| | - Walter R Bosch
- Department of Radiation Oncology, Washington University, St. Louis, MO, USA
| | - Christina Tsien
- Department of Radiation Oncology, McGill University Health Center, Montreal, QC, Canada
| | - Murat Surucu
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Julianne M Pollard-Larkin
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jan Schuemann
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Eduardo G Moros
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | | | - David J Gladstone
- Department of Radiation Oncology, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Heng Li
- Department of Radiation Oncology, Johns Hopkins University, Baltimore, MD, USA
| | - Charles B Simone
- Department of Radiation Oncology, New York Proton Center, New York, NY, USA
| | - Kristoffer Petersson
- Department of Radiation Oncology, MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | - Stephen F Kry
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amit Maity
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Billy W Loo
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lei Dong
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Peter G Maxim
- Department of Radiation Oncology, University of California Irvine, Irvine, CA, USA
| | - Ying Xiao
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeffrey C Buchsbaum
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| |
Collapse
|
46
|
Atkinson J, Bezak E, Le H, Kempson I. The current status of FLASH particle therapy: a systematic review. Phys Eng Sci Med 2023; 46:529-560. [PMID: 37160539 DOI: 10.1007/s13246-023-01266-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 04/20/2023] [Indexed: 05/11/2023]
Abstract
Particle therapies are becoming increasingly available clinically due to their beneficial energy deposition profile, sparing healthy tissues. This may be further promoted with ultra-high dose rates, termed FLASH. This review comprehensively summarises current knowledge based on studies relevant to proton- and carbon-FLASH therapy. As electron-FLASH literature presents important radiobiological findings that form the basis of proton and carbon-based FLASH studies, a summary of key electron-FLASH papers is also included. Preclinical data suggest three key mechanisms by which proton and carbon-FLASH are able to reduce normal tissue toxicities compared to conventional dose rates, with equipotent, or enhanced, tumour kill efficacy. However, a degree of caution is needed in clinically translating these findings as: most studies use transmission and do not conform the Bragg peak to tumour volume; mechanistic understanding is still in its infancy; stringent verification of dosimetry is rarely provided; biological assays are prone to limitations which need greater acknowledgement.
Collapse
Affiliation(s)
- Jake Atkinson
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia, 5095, Australia
| | - Eva Bezak
- Cancer Research Institute, University of South Australia, Adelaide, South Australia, 5000, Australia
- Department of Physics, University of Adelaide, North Terrace, Adelaide, South Australia, 5000, Australia
| | - Hien Le
- Department of Radiation Oncology, Royal Adelaide Hospital, Adelaide, 5000, Australia
| | - Ivan Kempson
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia, 5095, Australia.
| |
Collapse
|
47
|
Cooper CR, Jones DJL, Jones GDD, Petersson K. Comet Assay Profiling of FLASH-Induced Damage: Mechanistic Insights into the Effects of FLASH Irradiation. Int J Mol Sci 2023; 24:7195. [PMID: 37108360 PMCID: PMC10138874 DOI: 10.3390/ijms24087195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/06/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Numerous studies have demonstrated the normal tissue-sparing effects of ultra-high dose rate 'FLASH' irradiation in vivo, with an associated reduction in damage burden being reported in vitro. Towards this, two key radiochemical mechanisms have been proposed: radical-radical recombination (RRR) and transient oxygen depletion (TOD), with both being proposed to lead to reduced levels of induced damage. Previously, we reported that FLASH induces lower levels of DNA strand break damage in whole-blood peripheral blood lymphocytes (WB-PBL) ex vivo, but our study failed to distinguish the mechanism(s) involved. A potential outcome of RRR is the formation of crosslink damage (particularly, if any organic radicals recombine), whilst a possible outcome of TOD is a more anoxic profile of induced damage resulting from FLASH. Therefore, the aim of the current study was to profile FLASH-induced damage via the Comet assay, assessing any DNA crosslink formation as a putative marker of RRR and/or anoxic DNA damage formation as an indicative marker of TOD, to determine the extent to which either mechanism contributes to the "FLASH effect". Following FLASH irradiation, we see no evidence of any crosslink formation; however, FLASH irradiation induces a more anoxic profile of induced damage, supporting the TOD mechanism. Furthermore, treatment of WB-PBLs pre-irradiation with BSO abrogates the reduced strand break damage burden mediated by FLASH exposures. In summary, we do not see any experimental evidence to support the RRR mechanism contributing to the reduced damage burden induced by FLASH. However, the observation of a greater anoxic profile of damage following FLASH irradiation, together with the BSO abrogation of the reduced strand break damage burden mediated by FLASH, lends further support to TOD being a driver of the reduced damage burden plus a change in the damage profile mediated by FLASH.
Collapse
Affiliation(s)
- Christian R. Cooper
- Leicester Cancer Research Centre, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester LE2 7LX, UK; (D.J.L.J.); (G.D.D.J.)
- MRC Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Donald J. L. Jones
- Leicester Cancer Research Centre, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester LE2 7LX, UK; (D.J.L.J.); (G.D.D.J.)
| | - George D. D. Jones
- Leicester Cancer Research Centre, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester Royal Infirmary, Leicester LE2 7LX, UK; (D.J.L.J.); (G.D.D.J.)
| | - Kristoffer Petersson
- MRC Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
- Department of Haematology, Oncology and Radiation Physics, Skåne University Hospital Lund University, 221 85 Lund, Sweden
| |
Collapse
|
48
|
Limoli CL, Vozenin MC. Reinventing Radiobiology in the Light of FLASH Radiotherapy. ANNUAL REVIEW OF CANCER BIOLOGY 2023; 7:1-21. [PMID: 39421564 PMCID: PMC11486513 DOI: 10.1146/annurev-cancerbio-061421-022217] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Ultrahigh-dose rate FLASH radiotherapy (FLASH-RT) is a potentially paradigm-shifting treatment modality that holds the promise of expanding the therapeutic index for nearly any cancer. At the heart of this exciting technology comes the capability to ameliorate major normal tissue complications without compromising the efficacy of tumor killing. This combination of benefits has now been termed the FLASH effect and relies on an in vivo validation to rigorously demonstrate the absence of normal tissue toxicity. The FLASH effect occurs when the overall irradiation time is extremely short (<500 ms), and in this review we attempt to understand how FLASH-RT can kill tumors but spare normal tissues-likely the single most pressing question confronting the field today.
Collapse
Affiliation(s)
- Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, California, USA
| | - Marie-Catherine Vozenin
- Laboratory of Radiation Oncology, Radiation Oncology Service and Oncology Department, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
49
|
Charyyev S, Chang CW, Zhu M, Lin L, Langen K, Dhabaan A. Characterization of 250 MeV Protons from the Varian ProBeam PBS System for FLASH Radiation Therapy. Int J Part Ther 2023; 9:279-289. [PMID: 37169007 PMCID: PMC10166018 DOI: 10.14338/ijpt-22-00027.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 01/24/2023] [Indexed: 03/05/2023] Open
Abstract
Shoot-through proton FLASH radiation therapy has been proposed where the highest energy is extracted from a cyclotron to maximize the dose rate (DR). Although our proton pencil beam scanning system can deliver 250 MeV (the highest energy), this energy is not used clinically, and as such, 250 MeV has yet to be characterized during clinical commissioning. We aim to characterize the 250-MeV proton beam from the Varian ProBeam system for FLASH and assess the usability of the clinical monitoring ionization chamber (MIC) for FLASH use. We measured the following data for beam commissioning: integral depth dose curve, spot sigma, and absolute dose. To evaluate the MIC, we measured output as a function of beam current. To characterize a 250 MeV FLASH beam, we measured (1) the central axis DR as a function of current and spot spacing and arrangement, (2) for a fixed spot spacing, the maximum field size that achieves FLASH DR (ie, > 40 Gy/s), and (3) DR reproducibility. All FLASH DR measurements were performed using an ion chamber for the absolute dose, and irradiation times were obtained from log files. We verified dose measurements using EBT-XD films and irradiation times using a fast, pixelated spectral detector. R90 and R80 from integral depth dose were 37.58 and 37.69 cm, and spot sigma at the isocenter were σx = 3.336 and σy = 3.332 mm, respectively. The absolute dose output was measured as 0.343 Gy*mm2/MU for the commissioning conditions. Output was stable for beam currents up to 15 nA and gradually increased to 12-fold for 115 nA. Dose and DR depended on beam current, spot spacing, and arrangement and could be reproduced with 6.4% and 4.2% variations, respectively. Although FLASH was achieved and the largest field size that delivers FLASH DR was determined as 35 × 35 mm2, the current MIC has DR dependence, and users should measure dose and DR independently each time for their FLASH applications.
Collapse
Affiliation(s)
- Serdar Charyyev
- Department of Radiation Oncology, Stanford University, Palo Alto, CA, USA
| | - Chih-Wei Chang
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Mingyao Zhu
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Liyong Lin
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Katja Langen
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Anees Dhabaan
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| |
Collapse
|
50
|
Kanouta E, Poulsen PR, Kertzscher G, Sitarz MK, Sørensen BS, Johansen JG. Time-resolved dose rate measurements in pencil beam scanning proton FLASH therapy with a fiber-coupled scintillator detector system. Med Phys 2022; 50:2450-2462. [PMID: 36508162 DOI: 10.1002/mp.16156] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/08/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The spatial and temporal dose rate distribution of pencil beam scanning (PBS) proton therapy is important in ultra-high dose rate (UHDR) or FLASH irradiations. Validation of the temporal structure of the dose rate is crucial for quality assurance and may be performed using detectors with high temporal resolution and large dynamic range. PURPOSE To provide time-resolved in vivo dose rate measurements using a scintillator-based detector during proton PBS pre-clinical mouse experiments with dose rates ranging from conventional to UHDR. METHODS All irradiations were performed at the entrance plateau of a 250 MeV PBS proton beam. A detector system with four fiber-coupled ZnSe:O inorganic scintillators and 20 μs temporal resolution was used for dose rate measurements. The system was first characterized in terms of precision and stem signal. The detector precision was determined through repeated irradiations with the same field. The stem signal contribution was quantified by irradiating two of the detector probes alongside a bare fiber (fiber without a coupled scintillator). Next, the detector system was calibrated against an ionization chamber (IC) with all four detector probes and the IC placed in a water bath at 2 cm depth. A scan pattern covering 9.6 × 9.6 cm was used. Multiple irradiations with different requested nozzle currents provided instantaneous dose rates at the detector positions in the range of 7-1270 Gy/s. The correspondence of the detector signal (in Volts) to the instantaneous dose rate (in Gy/s) was found. The instantaneous dose rate was calculated from the beam current and the spot-to-detector distance assuming a Gaussian beam profile at distances up to 8 mm from the spot. Afterwards, the calibrated system was used in vivo, in mouse experiments, where mouse legs were irradiated with a constant dose and varying field dose rates of 0.7-87.5 Gy/s. The instantaneous dose rate was measured for each delivered spot and the delivered dose was determined as the integrated instantaneous dose rate. The spot dose profile and PBS dose rate map were calculated. The dose contamination to neighbouring mice were measured together with the upper limit of the dose to the mouse body. RESULTS The detectors showed high precision with ≤0.4% fluctuations in the measured dose. The stem signal exceeded 10% for spots <5 mm from the optical fiber and >18 mm from the scintillator. It contributed up to 0.2% to the total dose, which was considered negligible. All four detectors showed a non-linear relation between signal and instantaneous dose rate, which was modelled with a polynomial response function. In the mouse experiments, the measured scintillator dose showed 1.8% fluctuations, independent of the field dose rate. The in vivo measured spot dose profile had tails that deviated from a Gaussian profile with measurable dose contributions from spots up to 85 mm from the detector. Neighbour mouse irradiation contributed ∼1% of the total mouse dose. The upper limit of the mouse body dose was 6% of the mouse leg dose. CONCLUSIONS A fiber-coupled inorganic scintillator-based detector system can provide high precision in vivo measurements of the instantaneous dose rate if correction for the non-linear dose rate dependency is applied.
Collapse
Affiliation(s)
- Eleni Kanouta
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Per Rugaard Poulsen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark.,Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Mateusz Krzysztof Sitarz
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Brita Singers Sørensen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark.,Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jacob Graversen Johansen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark.,Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|