1
|
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
|
2
|
Amit U, Uslu U, Verginadis II, Kim MM, Motlagh SAO, Diffenderfer ES, Assenmacher CA, Bicher S, Atoche SJ, Ben-Josef E, Young RM, June CH, Koumenis C. Proton radiation boosts the efficacy of mesothelin-targeting chimeric antigen receptor T cell therapy in pancreatic cancer. Proc Natl Acad Sci U S A 2024; 121:e2403002121. [PMID: 39047033 PMCID: PMC11294999 DOI: 10.1073/pnas.2403002121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents a challenge in oncology, with limited treatment options for advanced-stage patients. Chimeric antigen receptor T cell (CAR T) therapy targeting mesothelin (MSLN) shows promise, but challenges such as the hostile immunosuppressive tumor microenvironment (TME) hinder its efficacy. This study explores the synergistic potential of combining proton radiation therapy (RT) with MSLN-targeting CAR T therapy in a syngeneic PDAC model. Proton RT significantly increased MSLN expression in tumor cells and caused a significant increase in CAR T cell infiltration into tumors. The combination therapy reshaped the immunosuppressive TME, promoting antitumorigenic M1 polarized macrophages and reducing myeloid-derived suppressor cells (MDSC). In a flank PDAC model, the combination therapy demonstrated superior attenuation of tumor growth and improved survival compared to individual treatments alone. In an orthotopic PDAC model treated with image-guided proton RT, tumor growth was significantly reduced in the combination group compared to the RT treatment alone. Further, the combination therapy induced an abscopal effect in a dual-flank tumor model, with increased serum interferon-γ levels and enhanced proliferation of extratumoral CAR T cells. In conclusion, combining proton RT with MSLN-targeting CAR T therapy proves effective in modulating the TME, enhancing CAR T cell trafficking, and exerting systemic antitumor effects. Thus, this combinatorial approach could present a promising strategy for improving outcomes in unresectable PDAC.
Collapse
Affiliation(s)
- Uri Amit
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Department of Radiation Oncology, Tel Aviv Medical Center, Tel Aviv64239, Israel
| | - Ugur Uslu
- Department of Pathology and Laboratory Medicine, Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA19104
| | - Ioannis I. Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Michele M. Kim
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Seyyedeh Azar Oliaei Motlagh
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Eric S. Diffenderfer
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Charles-Antoine Assenmacher
- Department of Pathobiology, School of Veterinary Medicine, Comparative Pathology Core, University of Pennsylvania, Philadelphia, PA19104
| | - Sandra Bicher
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Sebastian J. Atoche
- Department of Pathology and Laboratory Medicine, Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA19104
| | - Edgar Ben-Josef
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Regina M. Young
- Department of Pathology and Laboratory Medicine, Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA19104
| | - Carl H. June
- Department of Pathology and Laboratory Medicine, Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA19104
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA19104
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| |
Collapse
|
3
|
Kim K, Kim MM, Skoufos G, Diffenderfer ES, Motlagh SAO, Kokkorakis M, Koliaki I, Morcos G, Shoniyozov K, Griffin J, Hatzigeorgiou AG, Metz JM, Lin A, Feigenberg SJ, Cengel KA, Ky B, Koumenis C, Verginadis II. FLASH Proton Radiation Therapy Mitigates Inflammatory and Fibrotic Pathways and Preserves Cardiac Function in a Preclinical Mouse Model of Radiation-Induced Heart Disease. Int J Radiat Oncol Biol Phys 2024; 119:1234-1247. [PMID: 38364948 PMCID: PMC11209795 DOI: 10.1016/j.ijrobp.2024.01.224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 01/12/2024] [Accepted: 01/28/2024] [Indexed: 02/18/2024]
Abstract
PURPOSE Studies during the past 9 years suggest that delivering radiation at dose rates exceeding 40 Gy/s, known as "FLASH" radiation therapy, enhances the therapeutic index of radiation therapy (RT) by decreasing normal tissue damage while maintaining tumor response compared with conventional (or standard) RT. This study demonstrates the cardioprotective benefits of FLASH proton RT (F-PRT) compared with standard (conventional) proton RT (S-PRT), as evidenced by reduced acute and chronic cardiac toxicities. METHODS AND MATERIALS Mice were imaged using cone beam computed tomography to precisely determine the heart's apex as the beam isocenter. Irradiation was conducted using a shoot-through technique with a 5-mm diameter circular collimator. Bulk RNA-sequencing was performed on nonirradiated samples, as well as apexes treated with F-PRT or S-PRT, at 2 weeks after a single 40 Gy dose. Inflammatory responses were assessed through multiplex cytokine/chemokine microbead assay and immunofluorescence analyses. Levels of perivascular fibrosis were quantified using Masson's Trichrome and Picrosirius red staining. Additionally, cardiac tissue functionality was evaluated by 2-dimensional echocardiograms at 8- and 30-weeks post-PRT. RESULTS Radiation damage was specifically localized to the heart's apex. RNA profiling of cardiac tissues treated with PRT revealed that S-PRT uniquely upregulated pathways associated with DNA damage response, induction of tumor necrosis factor superfamily, and inflammatory response, and F-PRT primarily affected cytoplasmic translation, mitochondrion organization, and adenosine triphosphate synthesis. Notably, F-PRT led to a milder inflammatory response, accompanied by significantly attenuated changes in transforming growth factor β1 and α smooth muscle actin levels. Critically, F-PRT decreased collagen deposition and better preserved cardiac functionality compared with S-PRT. CONCLUSIONS This study demonstrated that F-PRT reduces the induction of an inflammatory environment with lower expression of inflammatory cytokines and profibrotic factors. Importantly, the results indicate that F-PRT better preserves cardiac functionality, as confirmed by echocardiography analysis, while also mitigating the development of long-term fibrosis.
Collapse
Affiliation(s)
- Kyle Kim
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michele M Kim
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Giorgos Skoufos
- Department of Electrical & Computer Engineering, University of Thessaly, Greece; Hellenic Pasteur Institute, Athens, Greece
| | - Eric S Diffenderfer
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Seyyedeh Azar Oliaei Motlagh
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michail Kokkorakis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ilektra Koliaki
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - George Morcos
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Khayrullo Shoniyozov
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joanna Griffin
- Department of Medicine, Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Artemis G Hatzigeorgiou
- Department of Electrical & Computer Engineering, University of Thessaly, Greece; Hellenic Pasteur Institute, Athens, Greece; DIANA-Laboratory, Department of Computer Science and Biomedical Informatics, University of Thessaly, Thessaly, Greece
| | - James M Metz
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alexander Lin
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Steven J Feigenberg
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Keith A Cengel
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bonnie Ky
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Ioannis I Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
4
|
Chowdhury P, Velalopoulou A, Verginadis II, Morcos G, Loo PE, Kim MM, Motlagh SAO, Shoniyozov K, Diffenderfer ES, Ocampo EA, Putt M, Assenmacher CA, Radaelli E, Lu J, Qin L, Liu H, Leli NM, Girdhani S, Denef N, Vander Stappen F, Cengel KA, Busch TM, Metz JM, Dong L, Lin A, Koumenis C. Proton FLASH Radiotherapy Ameliorates Radiation-induced Salivary Gland Dysfunction and Oral Mucositis and Increases Survival in a Mouse Model of Head and Neck Cancer. Mol Cancer Ther 2024; 23:877-889. [PMID: 38593239 PMCID: PMC11148539 DOI: 10.1158/1535-7163.mct-23-0663] [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: 09/26/2023] [Revised: 01/16/2024] [Accepted: 03/28/2024] [Indexed: 04/11/2024]
Abstract
Head and neck cancer radiotherapy often damages salivary glands and oral mucosa, severely negatively impacting patients' quality of life. The ability of FLASH proton radiotherapy (F-PRT) to decrease normal tissue toxicity while maintaining tumor control compared with standard proton radiotherapy (S-PRT) has been previously demonstrated for several tissues. However, its potential in ameliorating radiation-induced salivary gland dysfunction and oral mucositis and controlling orthotopic head and neck tumor growth has not been reported. The head and neck area of C57BL/6 mice was irradiated with a single dose of radiotherapy (ranging from 14-18 Gy) or a fractionated dose of 8 Gy × 3 of F-PRT (128 Gy/second) or S-PRT (0.95 Gy/second). Following irradiation, the mice were studied for radiation-induced xerostomia by measuring their salivary flow. Oral mucositis was analyzed by histopathologic examination. To determine the ability of F-PRT to control orthotopic head and neck tumors, tongue tumors were generated in the mice and then irradiated with either F-PRT or S-PRT. Mice treated with either a single dose or fractionated dose of F-PRT showed significantly improved survival than those irradiated with S-PRT. F-PRT-treated mice showed improvement in their salivary flow. S-PRT-irradiated mice demonstrated increased fibrosis in their tongue epithelium. F-PRT significantly increased the overall survival of the mice with orthotopic tumors compared with the S-PRT-treated mice. The demonstration that F-PRT decreases radiation-induced normal tissue toxicity without compromising tumor control, suggests that this modality could be useful for the clinical management of patients with head and neck cancer.
Collapse
Affiliation(s)
- Priyanka Chowdhury
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anastasia Velalopoulou
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ioannis I Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - George Morcos
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Phoebe E Loo
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Stanford University, Stanford, California
| | - Michele M Kim
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Seyyedeh Azar Oliaei Motlagh
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Khayrullo Shoniyozov
- 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
| | - Emilio A Ocampo
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- University of California, Los Angeles, California
| | - Mary Putt
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Charles-Antoine Assenmacher
- Penn Vet comparative Pathology Core, Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Enrico Radaelli
- Penn Vet comparative Pathology Core, Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jiawei Lu
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ling Qin
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hengxi Liu
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nektaria Maria Leli
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Swati Girdhani
- IBA (Ion Beam Applications S.A.), Ottignies-Louvain-la-Neuve, Belgium
| | - Nicolas Denef
- IBA (Ion Beam Applications S.A.), Ottignies-Louvain-la-Neuve, Belgium
| | | | - Keith A Cengel
- 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
| | - James M Metz
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lei Dong
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alexander Lin
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
5
|
Schneider M, Schilz JD, Schürer M, Gantz S, Dreyer A, Rothe G, Tillner F, Bodenstein E, Horst F, Beyreuther E. SAPPHIRE -establishment of small animal proton and photon image-guided radiation experiments. Phys Med Biol 2024; 69:095020. [PMID: 38537301 DOI: 10.1088/1361-6560/ad3887] [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: 01/17/2024] [Accepted: 03/26/2024] [Indexed: 04/25/2024]
Abstract
Thein vivoevolution of radiotherapy necessitates innovative platforms for preclinical investigation, bridging the gap between bench research and clinical applications. Understanding the nuances of radiation response, specifically tailored to proton and photon therapies, is critical for optimizing treatment outcomes. Within this context, preclinicalin vivoexperimental setups incorporating image guidance for both photon and proton therapies are pivotal, enabling the translation of findings from small animal models to clinical settings. TheSAPPHIREproject represents a milestone in this pursuit, presenting the installation of the small animal radiation therapy integrated beamline (SmART+ IB, Precision X-Ray Inc., Madison, Connecticut, USA) designed for preclinical image-guided proton and photon therapy experiments at University Proton Therapy Dresden. Through Monte Carlo simulations, low-dose on-site cone beam computed tomography imaging and quality assurance alignment protocols, the project ensures the safe and precise application of radiation, crucial for replicating clinical scenarios in small animal models. The creation of Hounsfield lookup tables and comprehensive proton and photon beam characterizations within this system enable accurate dose calculations, allowing for targeted and controlled comparison experiments. By integrating these capabilities,SAPPHIREbridges preclinical investigations and potential clinical applications, offering a platform for translational radiobiology research and cancer therapy advancements.
Collapse
Affiliation(s)
- Moritz Schneider
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiation Physics, Dresden, Germany
| | - Joshua D Schilz
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiation Physics, Dresden, Germany
| | - Michael Schürer
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Medizinische Fakultät and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Sebastian Gantz
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, Dresden, Germany
| | - Anne Dreyer
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, Dresden, Germany
| | - Gert Rothe
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Falk Tillner
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Elisabeth Bodenstein
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, Dresden, Germany
| | - Felix Horst
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, Dresden, Germany
| | - Elke Beyreuther
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiation Physics, Dresden, Germany
| |
Collapse
|
6
|
Evin M, Koumeir C, Bongrand A, Delpon G, Haddad F, Mouchard Q, Potiron V, Saade G, Servagent N, Villoing D, Métivier V, Chiavassa S. Methodology for small animals targeted irradiations at conventional and ultra-high dose rates 65 MeV proton beam. Phys Med 2024; 120:103332. [PMID: 38518627 DOI: 10.1016/j.ejmp.2024.103332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/20/2024] [Accepted: 03/11/2024] [Indexed: 03/24/2024] Open
Abstract
As part of translational research projects, mice may be irradiated on radiobiology platforms such as the one at the ARRONAX cyclotron. Generally, these platforms do not feature an integrated imaging system. Moreover, in the context of ultra-high dose-rate radiotherapy (FLASH-RT), treatment planning should consider potential changes in the beam characteristics and internal movements in the animal. A patient-like set-up and methodology has been implemented to ensure target coverage during conformal irradiations of the brain, lungs and intestines. In addition, respiratory cycle amplitudes were quantified by fluoroscopic acquisitions on a mouse, to ensure organ coverage and to assess the impact of respiration during FLASH-RT using the 4D digital phantom MOBY. Furthermore, beam incidence direction was studied from mice µCBCT and Monte Carlo simulations. Finally,in vivodosimetry with dose-rate independent radiochromic films (OC-1) and their LET dependency were investigated. The immobilization system ensures that the animal is held in a safe and suitable position. The geometrical evaluation of organ coverage, after the addition of the margins around the organs, was satisfactory. Moreover, no measured differences were found between CONV and FLASH beams enabling a single model of the beamline for all planning studies. Finally, the LET-dependency of the OC-1 film was determined and experimentally verified with phantoms, as well as the feasibility of using these filmsin vivoto validate the targeting. The methodology developed ensures accurate and reproducible preclinical irradiations in CONV and FLASH-RT without in-room image guidance in terms of positioning, dose calculation andin vivodosimetry.
Collapse
Affiliation(s)
- Manon Evin
- Nantes Université, IMT Atlantique, CNRS/IN2P3, SUBATECH, F-44000 Nantes, France.
| | - Charbel Koumeir
- Nantes Université, IMT Atlantique, CNRS/IN2P3, SUBATECH, F-44000 Nantes, France; GIP ARRONAX, Saint-Herblain, France
| | - Arthur Bongrand
- GIP ARRONAX, Saint-Herblain, France; Institut de Cancérologie de l'Ouest, site de Saint-Herblain, France
| | - Gregory Delpon
- Nantes Université, IMT Atlantique, CNRS/IN2P3, SUBATECH, F-44000 Nantes, France; Institut de Cancérologie de l'Ouest, site de Saint-Herblain, France
| | - Ferid Haddad
- Nantes Université, IMT Atlantique, CNRS/IN2P3, SUBATECH, F-44000 Nantes, France; GIP ARRONAX, Saint-Herblain, France
| | - Quentin Mouchard
- Nantes Université, IMT Atlantique, CNRS/IN2P3, SUBATECH, F-44000 Nantes, France
| | - Vincent Potiron
- Institut de Cancérologie de l'Ouest, site de Saint-Herblain, France; Nantes Université, CNRS, US2B, UMR 6286, F-44000 Nantes, France
| | - Gaëlle Saade
- Nantes Université, CNRS, US2B, UMR 6286, F-44000 Nantes, France
| | - Noël Servagent
- Nantes Université, IMT Atlantique, CNRS/IN2P3, SUBATECH, F-44000 Nantes, France
| | - Daphnée Villoing
- Institut de Cancérologie de l'Ouest, site de Saint-Herblain, France
| | - Vincent Métivier
- Nantes Université, IMT Atlantique, CNRS/IN2P3, SUBATECH, F-44000 Nantes, France
| | - Sophie Chiavassa
- Nantes Université, IMT Atlantique, CNRS/IN2P3, SUBATECH, F-44000 Nantes, France; Institut de Cancérologie de l'Ouest, site de Saint-Herblain, France
| |
Collapse
|
7
|
Reimold M, Assenbaum S, Bernert C, Beyreuther E, Brack FE, Karsch L, Kraft SD, Kroll F, Nossula A, Pawelke J, Rehwald M, Schlenvoigt HP, Schramm U, Umlandt MEP, Zeil K, Ziegler T, Metzkes-Ng J. Dosimetry for radiobiological in vivoexperiments at laser plasma-based proton accelerators. Phys Med Biol 2023; 68:185009. [PMID: 37579761 DOI: 10.1088/1361-6560/acf025] [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: 01/04/2023] [Accepted: 08/14/2023] [Indexed: 08/16/2023]
Abstract
Objective.Laser plasma-based accelerators (LPAs) of protons can contribute to research of ultra-high dose rate radiobiology as they provide pulse dose rates unprecedented at medical proton sources. Yet, LPAs pose challenges regarding precise and accurate dosimetry due to the high pulse dose rates, but also due to the sources' lower spectral stability and pulsed operation mode. Forin vivomodels, further challenges arise from the necessary small field dosimetry for volumetric dose distributions. For these novel source parameters and intended applications, a dosimetric standard needs to be established.Approach.In this work, we present a dosimetry and beam monitoring framework forin vivoirradiations of small target volumes with LPA protons, solving aforementioned challenges. The volumetric dose distribution in a sample (mean dose value and lateral/depth dose inhomogeneity) is provided by combining two independent dose measurements using radiochromic films (dose rate-independent) and ionization chambers (dose rate-dependent), respectively. The unique feature of the dosimetric setup is beam monitoring with a transmission time-of-flight spectrometer to quantify spectral fluctuations of the irradiating proton pulses. The resulting changes in the depth dose profile during irradiation of anin vivosample are hence accessible and enable pulse-resolved depth dose correction for each dose measurement.Main results.A first successful small animal pilot study using an LPA proton source serves as a testcase for the presented dosimetry approach and proves its performance in a realistic setting.Significance.With several facilities worldwide either setting up or already using LPA infrastructure for radiobiological studies with protons, the importance of LPA-adapted dosimetric frameworks as presented in this work is clearly underlined.
Collapse
Affiliation(s)
- Marvin Reimold
- Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany
- Technische Universität Dresden, D-01062 Dresden, Germany
| | - Stefan Assenbaum
- Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany
- Technische Universität Dresden, D-01062 Dresden, Germany
| | - Constantin Bernert
- Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany
- Technische Universität Dresden, D-01062 Dresden, Germany
| | - Elke Beyreuther
- Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany
- OncoRay-National Center for Radiation Research in Oncology, D-01309 Dresden, Germany
| | | | - Leonhard Karsch
- OncoRay-National Center for Radiation Research in Oncology, D-01309 Dresden, Germany
| | - Stephan D Kraft
- Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany
| | - Florian Kroll
- Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany
| | - Alexej Nossula
- Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany
- Martin-Luther-Universität Halle-Wittenberg, D-06120 Halle, Germany
| | - Jörg Pawelke
- Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany
- OncoRay-National Center for Radiation Research in Oncology, D-01309 Dresden, Germany
| | - Martin Rehwald
- Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany
| | | | - Ulrich Schramm
- Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany
- Technische Universität Dresden, D-01062 Dresden, Germany
| | - Marvin E P Umlandt
- Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany
- Technische Universität Dresden, D-01062 Dresden, Germany
| | - Karl Zeil
- Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany
| | - Tim Ziegler
- Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany
- Technische Universität Dresden, D-01062 Dresden, Germany
| | | |
Collapse
|
8
|
Palaniappan P, Knudsen Y, Meyer S, Gianoli C, Schnürle K, Würl M, Bortfeldt J, Parodi K, Riboldi M. Multi-stage image registration based on list-mode proton radiographies for small animal proton irradiation: A simulation study. Z Med Phys 2023:S0939-3889(23)00045-4. [PMID: 37353464 DOI: 10.1016/j.zemedi.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 03/27/2023] [Accepted: 04/03/2023] [Indexed: 06/25/2023]
Abstract
We present a multi-stage and multi-resolution deformable image registration framework for image-guidance at a small animal proton irradiation platform. The framework is based on list-mode proton radiographies acquired at different angles, which are used to deform a 3D treatment planning CT relying on normalized mutual information (NMI) or root mean square error (RMSE) in the projection domain. We utilized a mouse X-ray micro-CT expressed in relative stopping power (RSP), and obtained Monte Carlo simulations of proton images in list-mode for three different treatment sites (brain, head and neck, lung). Rigid transformations and controlled artificial deformation were applied to mimic position misalignments, weight loss and breathing changes. Results were evaluated based on the residual RMSE of RSP in the image domain including the comparison of extracted local features, i.e. between the reference micro-CT and the one transformed taking into account the calculated deformation. The residual RMSE of the RSP showed that the accuracy of the registration framework is promising for compensating rigid (>97% accuracy) and non-rigid (∼95% accuracy) transformations with respect to a conventional 3D-3D registration. Results showed that the registration accuracy is degraded when considering the realistic detector performance and NMI as a metric, whereas the RMSE in projection domain is rather insensitive. This work demonstrates the pre-clinical feasibility of the registration framework on different treatment sites and its use for small animal imaging with a realistic detector. Further computational optimization of the framework is required to enable the use of this tool for online estimation of the deformation.
Collapse
Affiliation(s)
- Prasannakumar Palaniappan
- Department of Medical Physics - Experimental Physics, Ludwig-Maximilians-Universität München, Munich, Germany.
| | - Yana Knudsen
- Department of Medical Physics - Experimental Physics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Sebastian Meyer
- Department of Medical Physics - Experimental Physics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Chiara Gianoli
- Department of Medical Physics - Experimental Physics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Katrin Schnürle
- Department of Medical Physics - Experimental Physics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Matthias Würl
- Department of Medical Physics - Experimental Physics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Jonathan Bortfeldt
- Department of Medical Physics - Experimental Physics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Katia Parodi
- Department of Medical Physics - Experimental Physics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Marco Riboldi
- Department of Medical Physics - Experimental Physics, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
9
|
Verhaegen F, Butterworth KT, Chalmers AJ, Coppes RP, de Ruysscher D, Dobiasch S, Fenwick JD, Granton PV, Heijmans SHJ, Hill MA, Koumenis C, Lauber K, Marples B, Parodi K, Persoon LCGG, Staut N, Subiel A, Vaes RDW, van Hoof S, Verginadis IL, Wilkens JJ, Williams KJ, Wilson GD, Dubois LJ. Roadmap for precision preclinical x-ray radiation studies. Phys Med Biol 2023; 68:06RM01. [PMID: 36584393 DOI: 10.1088/1361-6560/acaf45] [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: 05/12/2022] [Accepted: 12/30/2022] [Indexed: 12/31/2022]
Abstract
This Roadmap paper covers the field of precision preclinical x-ray radiation studies in animal models. It is mostly focused on models for cancer and normal tissue response to radiation, but also discusses other disease models. The recent technological evolution in imaging, irradiation, dosimetry and monitoring that have empowered these kinds of studies is discussed, and many developments in the near future are outlined. Finally, clinical translation and reverse translation are discussed.
Collapse
Affiliation(s)
- Frank Verhaegen
- MAASTRO Clinic, Radiotherapy Division, GROW-School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
- SmART Scientific Solutions BV, Maastricht, The Netherlands
| | - Karl T Butterworth
- Patrick G. Johnston, Centre for Cancer Research, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Anthony J Chalmers
- School of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, United Kingdom
| | - Rob P Coppes
- Departments of Biomedical Sciences of Cells & Systems, Section Molecular Cell Biology and Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands
| | - Dirk de Ruysscher
- MAASTRO Clinic, Radiotherapy Division, GROW-School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Sophie Dobiasch
- Department of Radiation Oncology, Technical University of Munich (TUM), School of Medicine and Klinikum rechts der Isar, Germany
- Department of Medical Physics, Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, Germany
| | - John D Fenwick
- Department of Medical Physics & Biomedical Engineering University College LondonMalet Place Engineering Building, London WC1E 6BT, United Kingdom
| | | | | | - Mark A Hill
- MRC Oxford Institute for Radiation Oncology, University of Oxford, ORCRB Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kirsten Lauber
- Department of Radiation Oncology, University Hospital, LMU München, Munich, Germany
- German Cancer Consortium (DKTK), Partner site Munich, Germany
| | - Brian Marples
- Department of Radiation Oncology, University of Rochester, NY, United States of America
| | - Katia Parodi
- German Cancer Consortium (DKTK), Partner site Munich, Germany
- Department of Medical Physics, Faculty of Physics, Ludwig-Maximilians-Universität München, Garching b. Munich, Germany
| | | | - Nick Staut
- SmART Scientific Solutions BV, Maastricht, The Netherlands
| | - Anna Subiel
- National Physical Laboratory, Medical Radiation Science Hampton Road, Teddington, Middlesex, TW11 0LW, United Kingdom
| | - Rianne D W Vaes
- MAASTRO Clinic, Radiotherapy Division, GROW-School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | | | - Ioannis L Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jan J Wilkens
- Department of Radiation Oncology, Technical University of Munich (TUM), School of Medicine and Klinikum rechts der Isar, Germany
- Physics Department, Technical University of Munich (TUM), Germany
| | - Kaye J Williams
- Division of Pharmacy and Optometry, University of Manchester, Manchester, United Kingdom
| | - George D Wilson
- Department of Radiation Oncology, Beaumont Health, MI, United States of America
- Henry Ford Health, Detroit, MI, United States of America
| | - Ludwig J Dubois
- The M-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
10
|
Schneider M, Bodenstein E, Bock J, Dietrich A, Gantz S, Heuchel L, Krause M, Lühr A, von Neubeck C, Nexhipi S, Schürer M, Tillner F, Beyreuther E, Suckert T, Müller JR. Combined proton radiography and irradiation for high-precision preclinical studies in small animals. Front Oncol 2022; 12:982417. [PMID: 36419890 PMCID: PMC9677333 DOI: 10.3389/fonc.2022.982417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/02/2022] [Indexed: 08/22/2023] Open
Abstract
BACKGROUND AND PURPOSE Proton therapy has become a popular treatment modality in the field of radiooncology due to higher spatial dose conformity compared to conventional radiotherapy, which holds the potential to spare normal tissue. Nevertheless, unresolved research questions, such as the much debated relative biological effectiveness (RBE) of protons, call for preclinical research, especially regarding in vivo studies. To mimic clinical workflows, high-precision small animal irradiation setups with image-guidance are needed. MATERIAL AND METHODS A preclinical experimental setup for small animal brain irradiation using proton radiographies was established to perform planning, repositioning, and irradiation of mice. The image quality of proton radiographies was optimized regarding the resolution, contrast-to-noise ratio (CNR), and minimal dose deposition in the animal. Subsequently, proof-of-concept histological analysis was conducted by staining for DNA double-strand breaks that were then correlated to the delivered dose. RESULTS The developed setup and workflow allow precise brain irradiation with a lateral target positioning accuracy of<0.26mm for in vivo experiments at minimal imaging dose of<23mGy per mouse. The custom-made software for image registration enables the fast and precise animal positioning at the beam with low observer-variability. DNA damage staining validated the successful positioning and irradiation of the mouse hippocampus. CONCLUSION Proton radiography enables fast and effective high-precision lateral alignment of proton beam and target volume in mouse irradiation experiments with limited dose exposure. In the future, this will enable irradiation of larger animal cohorts as well as fractionated proton irradiation.
Collapse
Affiliation(s)
- Moritz Schneider
- OncoRay, National Center for Radiation Research in Oncology- Faculty of Medicine and University Hospital Carl Gustav Carus- Technische Universitat Dresden-Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiation Physics, Dresden, Germany
| | - Elisabeth Bodenstein
- OncoRay, National Center for Radiation Research in Oncology- Faculty of Medicine and University Hospital Carl Gustav Carus- Technische Universitat Dresden-Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Johanna Bock
- OncoRay, National Center for Radiation Research in Oncology- Faculty of Medicine and University Hospital Carl Gustav Carus- Technische Universitat Dresden-Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Antje Dietrich
- OncoRay, National Center for Radiation Research in Oncology- Faculty of Medicine and University Hospital Carl Gustav Carus- Technische Universitat Dresden-Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- German Cancer Consortium Deutsches Konsortium für Translationale Krebsforschung (DKTK), partner site Dresden- German Cancer Research Center DKFZ, Heidelberg, Germany
| | - Sebastian Gantz
- OncoRay, National Center for Radiation Research in Oncology- Faculty of Medicine and University Hospital Carl Gustav Carus- Technische Universitat Dresden-Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Lena Heuchel
- Technical University (TU) Dortmund- Faculty of Physics, Medical Physics and Radiotherapy, Dortmund, Germany
| | - Mechthild Krause
- OncoRay, National Center for Radiation Research in Oncology- Faculty of Medicine and University Hospital Carl Gustav Carus- Technische Universitat Dresden-Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
- German Cancer Consortium Deutsches Konsortium für Translationale Krebsforschung (DKTK), partner site Dresden- German Cancer Research Center DKFZ, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universitat Dresden, Dresden, Germany
| | - Armin Lühr
- OncoRay, National Center for Radiation Research in Oncology- Faculty of Medicine and University Hospital Carl Gustav Carus- Technische Universitat Dresden-Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
- Technical University (TU) Dortmund- Faculty of Physics, Medical Physics and Radiotherapy, Dortmund, Germany
| | - Cläre von Neubeck
- OncoRay, National Center for Radiation Research in Oncology- Faculty of Medicine and University Hospital Carl Gustav Carus- Technische Universitat Dresden-Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- German Cancer Consortium Deutsches Konsortium für Translationale Krebsforschung (DKTK), partner site Dresden- German Cancer Research Center DKFZ, Heidelberg, Germany
- Department of Particle Therapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sindi Nexhipi
- OncoRay, National Center for Radiation Research in Oncology- Faculty of Medicine and University Hospital Carl Gustav Carus- Technische Universitat Dresden-Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Michael Schürer
- OncoRay, National Center for Radiation Research in Oncology- Faculty of Medicine and University Hospital Carl Gustav Carus- Technische Universitat Dresden-Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
| | - Falk Tillner
- OncoRay, National Center for Radiation Research in Oncology- Faculty of Medicine and University Hospital Carl Gustav Carus- Technische Universitat Dresden-Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universitat Dresden, Dresden, Germany
| | - Elke Beyreuther
- OncoRay, National Center for Radiation Research in Oncology- Faculty of Medicine and University Hospital Carl Gustav Carus- Technische Universitat Dresden-Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiation Physics, Dresden, Germany
| | - Theresa Suckert
- OncoRay, National Center for Radiation Research in Oncology- Faculty of Medicine and University Hospital Carl Gustav Carus- Technische Universitat Dresden-Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- German Cancer Consortium Deutsches Konsortium für Translationale Krebsforschung (DKTK), partner site Dresden- German Cancer Research Center DKFZ, Heidelberg, Germany
| | - Johannes Richard Müller
- OncoRay, National Center for Radiation Research in Oncology- Faculty of Medicine and University Hospital Carl Gustav Carus- Technische Universitat Dresden-Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Deutsche Forschungsgemeinschaft Cluster of Excellence 'Physics of Life', Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
11
|
Comparing radiolytic production of H 2O 2 and development of Zebrafish embryos after ultra high dose rate exposure with electron and transmission proton beams. Radiother Oncol 2022; 175:197-202. [PMID: 35868604 DOI: 10.1016/j.radonc.2022.07.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/30/2022] [Accepted: 07/13/2022] [Indexed: 11/21/2022]
Abstract
The physico-chemical and biological response to conventional and UHDR electron and proton beams was investigated, along with conventional photons. The temporal structure and nature of the beam affected both, with electron beam at ≥1400 Gy/s and proton beam at 0.1 and 1260 Gy/s found to be isoefficient at sparing zebrafish embryos.
Collapse
|
12
|
Kim MM, Darafsheh A, Schuemann J, Dokic I, Lundh O, Zhao T, Ramos-Méndez J, Dong L, Petersson K. Development of Ultra-High Dose-Rate (FLASH) Particle Therapy. IEEE TRANSACTIONS ON RADIATION AND PLASMA MEDICAL SCIENCES 2022; 6:252-262. [PMID: 36092270 PMCID: PMC9457346 DOI: 10.1109/trpms.2021.3091406] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Research efforts in FLASH radiotherapy have increased at an accelerated pace recently. FLASH radiotherapy involves ultra-high dose rates and has shown to reduce toxicity to normal tissue while maintaining tumor response in pre-clinical studies when compared to conventional dose rate radiotherapy. The goal of this review is to summarize the studies performed to-date with proton, electron, and heavy ion FLASH radiotherapy, with particular emphasis on the physical aspects of each study and the advantages and disadvantages of each modality. Beam delivery parameters, experimental set-up, and the dosimetry tools used are described for each FLASH modality. In addition, modeling efforts and treatment planning for FLASH radiotherapy is discussed along with potential drawbacks when translated into the clinical setting. The final section concludes with further questions that have yet to be answered before safe clinical implementation of FLASH radiotherapy.
Collapse
Affiliation(s)
- Michele M Kim
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Arash Darafsheh
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jan Schuemann
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ivana Dokic
- Clinical Cooperation Unit Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460, Heidelberg, Germany
- Division of Molecular and Translational Radiation Oncology, Department of Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), Im Neuenheimer Feld 450, 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK) Core-Center Heidelberg, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Oncology (NCRO), Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 222, Heidelberg, Germany
| | - Olle Lundh
- Department of Physics, Lund University, Lund, Sweden
| | - Tianyu Zhao
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - José Ramos-Méndez
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
| | - Lei Dong
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kristoffer Petersson
- Department of Oncology, The Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
- Radiation Physics, Department of Haematology, Oncology and Radiation Physics, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
13
|
Malimban J, Lathouwers D, Qian H, Verhaegen F, Wiedemann J, Brandenburg S, Staring M. Deep learning-based segmentation of the thorax in mouse micro-CT scans. Sci Rep 2022; 12:1822. [PMID: 35110676 PMCID: PMC8810936 DOI: 10.1038/s41598-022-05868-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 01/18/2022] [Indexed: 12/18/2022] Open
Abstract
For image-guided small animal irradiations, the whole workflow of imaging, organ contouring, irradiation planning, and delivery is typically performed in a single session requiring continuous administration of anaesthetic agents. Automating contouring leads to a faster workflow, which limits exposure to anaesthesia and thereby, reducing its impact on experimental results and on animal wellbeing. Here, we trained the 2D and 3D U-Net architectures of no-new-Net (nnU-Net) for autocontouring of the thorax in mouse micro-CT images. We trained the models only on native CTs and evaluated their performance using an independent testing dataset (i.e., native CTs not included in the training and validation). Unlike previous studies, we also tested the model performance on an external dataset (i.e., contrast-enhanced CTs) to see how well they predict on CTs completely different from what they were trained on. We also assessed the interobserver variability using the generalized conformity index ([Formula: see text]) among three observers, providing a stronger human baseline for evaluating automated contours than previous studies. Lastly, we showed the benefit on the contouring time compared to manual contouring. The results show that 3D models of nnU-Net achieve superior segmentation accuracy and are more robust to unseen data than 2D models. For all target organs, the mean surface distance (MSD) and the Hausdorff distance (95p HD) of the best performing model for this task (nnU-Net 3d_fullres) are within 0.16 mm and 0.60 mm, respectively. These values are below the minimum required contouring accuracy of 1 mm for small animal irradiations, and improve significantly upon state-of-the-art 2D U-Net-based AIMOS method. Moreover, the conformity indices of the 3d_fullres model also compare favourably to the interobserver variability for all target organs, whereas the 2D models perform poorly in this regard. Importantly, the 3d_fullres model offers 98% reduction in contouring time.
Collapse
Affiliation(s)
- Justin Malimban
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands.
| | - Danny Lathouwers
- Department of Radiation Science and Technology, Faculty of Applied Sciences, Delft University of Technology, 2629 JB, Delft, The Netherlands
| | - Haibin Qian
- Department of Medical Biology, Amsterdam University Medical Centers (Location AMC) and Cancer Center Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Frank Verhaegen
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, 6229 ER, Maastricht, The Netherlands
| | - Julia Wiedemann
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands
- Department of Biomedical Sciences of Cells and Systems-Section Molecular Cell Biology, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands
| | - Sytze Brandenburg
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands
| | - Marius Staring
- Department of Radiology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| |
Collapse
|
14
|
Vorhees CV, Vatner RE, Williams MT. Review of Conventional and High Dose Rate Brain Radiation (FLASH): Neurobehavioural, Neurocognitive and Assessment Issues in Rodent Models. Clin Oncol (R Coll Radiol) 2021; 33:e482-e491. [PMID: 34548203 PMCID: PMC10114147 DOI: 10.1016/j.clon.2021.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/29/2021] [Accepted: 09/02/2021] [Indexed: 10/20/2022]
Abstract
Ionising radiation causes secondary tumours and/or enduring cognitive deficits, especially in children. Proton radiotherapy reduces exposure of the developing brain in children but may still cause some lasting effects. Recent observations show that ultra-high dose rate radiation treatment (≥40 Gy/s), called the FLASH effect, is equally effective at tumour control but less damaging to surrounding tissue compared with conventional dose rate protons (0.03-3 Gy/s). Most studies on the FLASH effect in brain and other tissues with different radiation modalities (electron and photon radiation), show FLASH benefits in these preclinical rodent models, but the data are limited, especially for proton FLASH, including for dose, dose rate and neurochemical and neurobehavioural outcomes. Tests of neurocognitive outcomes have been limited despite clinical evidence that this is the area of greatest concern. The FLASH effect in the context of proton exposure is promising, but a more systematic and comprehensive approach to outcomes is needed.
Collapse
Affiliation(s)
- C V Vorhees
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Cincinnati Children's/University of Cincinnati Proton Therapy and Research Center, Cincinnati, Ohio, USA.
| | - R E Vatner
- Cincinnati Children's/University of Cincinnati Proton Therapy and Research Center, Cincinnati, Ohio, USA; Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - M T Williams
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Cincinnati Children's/University of Cincinnati Proton Therapy and Research Center, Cincinnati, Ohio, USA
| |
Collapse
|
15
|
Diffenderfer ES, Sørensen BS, Mazal A, Carlson DJ. The current status of preclinical proton FLASH radiation and future directions. Med Phys 2021; 49:2039-2054. [PMID: 34644403 DOI: 10.1002/mp.15276] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 09/12/2021] [Accepted: 09/17/2021] [Indexed: 11/05/2022] Open
Abstract
We review the current status of proton FLASH experimental systems, including preclinical physical and biological results. Technological limitations on preclinical investigation of FLASH biological mechanisms and determination of clinically relevant parameters are discussed. A review of the biological data reveals no reproduced proton FLASH effect in vitro and a significant in vivo FLASH sparing effect of normal tissue toxicity observed with multiple proton FLASH irradiation systems. Importantly, multiple studies suggest little or no difference in tumor growth delay for proton FLASH when compared to conventional dose rate proton radiation. A discussion follows on future areas of development with a focus on the determination of the optimal parameters for maximizing the therapeutic ratio between tumor and normal tissue response and ultimately clinical translation of proton FLASH radiation.
Collapse
Affiliation(s)
- Eric S Diffenderfer
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Brita S Sørensen
- Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark.,Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Alejandro Mazal
- Department of Medical Physics, Centro de Protonterapia Quironsalud, Madrid, Spain
| | - David J Carlson
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
16
|
Comparison of FLASH Proton Entrance and the Spread-Out Bragg Peak Dose Regions in the Sparing of Mouse Intestinal Crypts and in a Pancreatic Tumor Model. Cancers (Basel) 2021; 13:cancers13164244. [PMID: 34439398 PMCID: PMC8392865 DOI: 10.3390/cancers13164244] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/14/2021] [Accepted: 08/19/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary FLASH radiotherapy is a treatment technique of interest that involves radiation delivered at ultra-high dose rates >100 times faster than traditional radiation therapy, which has been shown to spare radiation damage to normal tissue but maintain tumor control capabilities. Proton therapy uses spread-out proton Bragg peaks to reduce radiation dose to normal tissue by directing the highest dose of radiation to the tumor volume. In this study, irradiation of the whole abdomen of mice was performed with proton beams at FLASH dose rates in order to investigate the normal tissue sparing capabilities of the spread-out Bragg peak compared to the entrance region of the proton depth dose curve. Abstract Ultra-high dose rate FLASH proton radiotherapy (F-PRT) has been shown to reduce normal tissue toxicity compared to standard dose rate proton radiotherapy (S-PRT) in experiments using the entrance portion of the proton depth dose profile, while proton therapy uses a spread-out Bragg peak (SOBP) with unknown effects on FLASH toxicity sparing. To investigate, the biological effects of F-PRT using an SOBP and the entrance region were compared to S-PRT in mouse intestine. In this study, 8–10-week-old C57BL/6J mice underwent 15 Gy (absorbed dose) whole abdomen irradiation in four groups: (1) SOBP F-PRT, (2) SOBP S-PRT, (3) entrance F-PRT, and (4) entrance S-PRT. Mice were injected with EdU 3.5 days after irradiation, and jejunum segments were harvested and preserved. EdU-positive proliferating cells and regenerated intestinal crypts were quantified. The SOBP had a modulation (width) of 2.5 cm from the proximal to distal 90%. Dose rates with a SOBP for F-PRT or S-PRT were 108.2 ± 8.3 Gy/s or 0.82 ± 0.14 Gy/s, respectively. In the entrance region, dose rates were 107.1 ± 15.2 Gy/s and 0.83 ± 0.19 Gy/s, respectively. Both entrance and SOBP F-PRT preserved a significantly higher number of EdU + /crypt cells and percentage of regenerated crypts compared to S-PRT. Moreover, tumor growth studies showed no difference between SOBP and entrance for either of the treatment modalities.
Collapse
|
17
|
Suckert T, Nexhipi S, Dietrich A, Koch R, Kunz-Schughart LA, Bahn E, Beyreuther E. Models for Translational Proton Radiobiology-From Bench to Bedside and Back. Cancers (Basel) 2021; 13:4216. [PMID: 34439370 PMCID: PMC8395028 DOI: 10.3390/cancers13164216] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/09/2021] [Accepted: 08/17/2021] [Indexed: 12/25/2022] Open
Abstract
The number of proton therapy centers worldwide are increasing steadily, with more than two million cancer patients treated so far. Despite this development, pending questions on proton radiobiology still call for basic and translational preclinical research. Open issues are the on-going discussion on an energy-dependent varying proton RBE (relative biological effectiveness), a better characterization of normal tissue side effects and combination treatments with drugs originally developed for photon therapy. At the same time, novel possibilities arise, such as radioimmunotherapy, and new proton therapy schemata, such as FLASH irradiation and proton mini-beams. The study of those aspects demands for radiobiological models at different stages along the translational chain, allowing the investigation of mechanisms from the molecular level to whole organisms. Focusing on the challenges and specifics of proton research, this review summarizes the different available models, ranging from in vitro systems to animal studies of increasing complexity as well as complementing in silico approaches.
Collapse
Affiliation(s)
- Theresa Suckert
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (T.S.); (S.N.); (A.D.); (L.A.K.-S.)
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sindi Nexhipi
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (T.S.); (S.N.); (A.D.); (L.A.K.-S.)
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01309 Dresden, Germany
| | - Antje Dietrich
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (T.S.); (S.N.); (A.D.); (L.A.K.-S.)
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Robin Koch
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany; (R.K.); (E.B.)
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Leoni A. Kunz-Schughart
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (T.S.); (S.N.); (A.D.); (L.A.K.-S.)
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
| | - Emanuel Bahn
- Heidelberg Institute of Radiation Oncology (HIRO), 69120 Heidelberg, Germany; (R.K.); (E.B.)
- Department of Radiation Oncology, Heidelberg University Hospital, 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
- German Cancer Research Center (DKFZ), Clinical Cooperation Unit Radiation Oncology, 69120 Heidelberg, Germany
| | - Elke Beyreuther
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01309 Dresden, Germany; (T.S.); (S.N.); (A.D.); (L.A.K.-S.)
- Helmholtz-Zentrum Dresden—Rossendorf, Institute of Radiation Physics, 01328 Dresden, Germany
| |
Collapse
|
18
|
Vatner R, James CD, Sathiaseelan V, Bondra KM, Kalapurakal JA, Houghton PJ. Radiation therapy and molecular-targeted agents in preclinical testing for immunotherapy, brain tumors, and sarcomas: Opportunities and challenges. Pediatr Blood Cancer 2021; 68 Suppl 2:e28439. [PMID: 32827353 DOI: 10.1002/pbc.28439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 04/24/2020] [Accepted: 05/07/2020] [Indexed: 01/07/2023]
Abstract
Despite radiation therapy (RT) being an integral part of the treatment of most pediatric cancers and the recent discovery of novel molecular-targeted agents (MTAs) in this era of precision medicine with the potential to improve the therapeutic ratio of modern chemoradiotherapy regimens, there are only a few preclinical trials being conducted to discover novel radiosensitizers and radioprotectors. This has resulted in a paucity of translational clinical trials combining RT and novel MTAs. This report describes the opportunities and challenges of investigating RT together with MTAs in preclinical testing for immunotherapy, brain tumors, and sarcomas in pediatric oncology. We discuss the need for improving the collaboration between radiation oncologists, biologists, and physicists to improve the reliability, reproducibility, and translational potential of RT-based preclinical research. Current translational clinical trials using RT and MTAs for immunotherapy, brain tumors, and sarcomas are described. The technologic advances in experimental RT, availability of novel experimental tumor models, advances in immunology and tumor biology, and the discovery of novel MTAs together hold considerable promise for good quality preclinical and clinical multimodality research to improve the current rates of survival and toxicity in children afflicted with cancer.
Collapse
Affiliation(s)
- Ralph Vatner
- Radiation Oncology, University of Cincinnati and Cincinnati Children's Hospital, Cincinnati, Ohio
| | | | | | - Kathryn M Bondra
- Greehey Children's Cancer Research Institute, University of Texas, San Antonio, Texas
| | | | - Peter J Houghton
- Greehey Children's Cancer Research Institute, University of Texas, San Antonio, Texas
| |
Collapse
|
19
|
Ko RB, Soto LA, von Eyben R, Melemenidis S, Rankin EB, Maxim PG, Graves EE, Loo BW. Evaluating the Reproducibility of Mouse Anatomy under Rotation in a Custom Immobilization Device for Conformal FLASH Radiotherapy. Radiat Res 2021; 194:600-606. [PMID: 32857849 DOI: 10.1667/rade-20-00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 06/18/2020] [Indexed: 11/03/2022]
Abstract
The observation of an enhanced therapeutic index for FLASH radiotherapy in mice has created interest in practical laboratory-based FLASH irradiators. To date, systems capable of 3D conformal FLASH irradiation in mice have been lacking. We are developing such a system, incorporating a high-current linear accelerator to produce a collimated X-ray beam in a stationary beamline design, rotating the mouse about a longitudinal axis to achieve conformal irradiation from multiple beam directions. The purpose of this work was to evaluate the reproducibility of mouse anatomy under rotation at speeds compatible with conformal FLASH delivery. Three short-hair mice and two hairless mice were immobilized under anesthesia in body weight-specific contoured plastic molds, and subjected to three rotational (up to 3 revolutions/s) and two non-rotational movement interventions. MicroCT images were acquired before and after each intervention. The displacements of 11 anatomic landmarks were measured on the image pairs. The displacement of the anatomical landmarks with any of the interventions was 0.5 mm or less for 92.4% of measurements, with a single measurement out of 275 (11 landmarks × 5 interventions × 5 mice) reaching 1 mm. There was no significant difference in the displacements associated with rotation compared to those associated with moving the immobilized mouse in and out of a scanner or with leaving the mouse in place for 5 min with no motion. There were no significant differences in displacements between mice with or without hair, although the analysis is limited by small numbers, or between different anatomic landmarks. These results show that anatomic reproducibility under rotation speed corresponding to FLASH irradiation times appears to be compatible with conformal/stereotactic irradiation in mice.
Collapse
Affiliation(s)
- Ryan B Ko
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Luis A Soto
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Rie von Eyben
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Stavros Melemenidis
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - Erinn B Rankin
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California.,Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California.,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Peter G Maxim
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Edward E Graves
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California.,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Billy W Loo
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California.,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
20
|
Gerlach S, Pinto M, Kurichiyanil N, Grau C, Hérault J, Hillbrand M, Poulsen PR, Safai S, Schippers JM, Schwarz M, Søndergaard CS, Tommasino F, Verroi E, Vidal M, Yohannes I, Schreiber J, Parodi K. Beam characterization and feasibility study for a small animal irradiation platform at clinical proton therapy facilities. Phys Med Biol 2020; 65:245045. [DOI: 10.1088/1361-6560/abc832] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
21
|
Ostermayr TM, Kreuzer C, Englbrecht FS, Gebhard J, Hartmann J, Huebl A, Haffa D, Hilz P, Parodi K, Wenz J, Donovan ME, Dyer G, Gaul E, Gordon J, Martinez M, Mccary E, Spinks M, Tiwari G, Hegelich BM, Schreiber J. Laser-driven x-ray and proton micro-source and application to simultaneous single-shot bi-modal radiographic imaging. Nat Commun 2020; 11:6174. [PMID: 33268784 PMCID: PMC7710721 DOI: 10.1038/s41467-020-19838-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 10/29/2020] [Indexed: 11/16/2022] Open
Abstract
Radiographic imaging with x-rays and protons is an omnipresent tool in basic research and applications in industry, material science and medical diagnostics. The information contained in both modalities can often be valuable in principle, but difficult to access simultaneously. Laser-driven solid-density plasma-sources deliver both kinds of radiation, but mostly single modalities have been explored for applications. Their potential for bi-modal radiographic imaging has never been fully realized, due to problems in generating appropriate sources and separating image modalities. Here, we report on the generation of proton and x-ray micro-sources in laser-plasma interactions of the focused Texas Petawatt laser with solid-density, micrometer-sized tungsten needles. We apply them for bi-modal radiographic imaging of biological and technological objects in a single laser shot. Thereby, advantages of laser-driven sources could be enriched beyond their small footprint by embracing their additional unique properties, including the spectral bandwidth, small source size and multi-mode emission. Here the authors show a synchronized single-shot bi-modal x-ray and proton source based on laser-generated plasma. This source can be useful for radiographic and tomographic imaging.
Collapse
Affiliation(s)
- T M Ostermayr
- Ludwig-Maximilians-Universität München, Fakultät für Physik, 85748, Garching, Germany. .,Max-Planck-Institut für Quantenoptik, 85748, Garching, Germany. .,Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA.
| | - C Kreuzer
- Ludwig-Maximilians-Universität München, Fakultät für Physik, 85748, Garching, Germany
| | - F S Englbrecht
- Ludwig-Maximilians-Universität München, Fakultät für Physik, 85748, Garching, Germany
| | - J Gebhard
- Ludwig-Maximilians-Universität München, Fakultät für Physik, 85748, Garching, Germany
| | - J Hartmann
- Ludwig-Maximilians-Universität München, Fakultät für Physik, 85748, Garching, Germany
| | - A Huebl
- Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - D Haffa
- Ludwig-Maximilians-Universität München, Fakultät für Physik, 85748, Garching, Germany
| | - P Hilz
- Ludwig-Maximilians-Universität München, Fakultät für Physik, 85748, Garching, Germany.,Helmholtz Institute Jena, 07743, Jena, Germany
| | - K Parodi
- Ludwig-Maximilians-Universität München, Fakultät für Physik, 85748, Garching, Germany
| | - J Wenz
- Ludwig-Maximilians-Universität München, Fakultät für Physik, 85748, Garching, Germany
| | - M E Donovan
- Center for High Energy Density Science, University of Texas at Austin, Austin, TX, 78712, USA
| | - G Dyer
- Center for High Energy Density Science, University of Texas at Austin, Austin, TX, 78712, USA
| | - E Gaul
- Center for High Energy Density Science, University of Texas at Austin, Austin, TX, 78712, USA
| | - J Gordon
- Center for High Energy Density Science, University of Texas at Austin, Austin, TX, 78712, USA
| | - M Martinez
- Center for High Energy Density Science, University of Texas at Austin, Austin, TX, 78712, USA
| | - E Mccary
- Center for High Energy Density Science, University of Texas at Austin, Austin, TX, 78712, USA
| | - M Spinks
- Center for High Energy Density Science, University of Texas at Austin, Austin, TX, 78712, USA
| | - G Tiwari
- Center for High Energy Density Science, University of Texas at Austin, Austin, TX, 78712, USA
| | - B M Hegelich
- Center for High Energy Density Science, University of Texas at Austin, Austin, TX, 78712, USA
| | - J Schreiber
- Ludwig-Maximilians-Universität München, Fakultät für Physik, 85748, Garching, Germany. .,Max-Planck-Institut für Quantenoptik, 85748, Garching, Germany.
| |
Collapse
|
22
|
Diffenderfer ES, Verginadis II, Kim MM, Shoniyozov K, Velalopoulou A, Goia D, Putt M, Hagan S, Avery S, Teo K, Zou W, Lin A, Swisher-McClure S, Koch C, Kennedy AR, Minn A, Maity A, Busch TM, Dong L, Koumenis C, Metz J, Cengel KA. Design, Implementation, and in Vivo Validation of a Novel Proton FLASH Radiation Therapy System. Int J Radiat Oncol Biol Phys 2020; 106:440-448. [PMID: 31928642 DOI: 10.1016/j.ijrobp.2019.10.049] [Citation(s) in RCA: 273] [Impact Index Per Article: 68.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 10/11/2019] [Accepted: 10/31/2019] [Indexed: 11/25/2022]
Abstract
PURPOSE Recent studies suggest that ultrahigh-dose-rate, "FLASH," electron radiation therapy (RT) decreases normal tissue damage while maintaining tumor response compared with conventional dose rate RT. Here, we describe a novel RT apparatus that delivers FLASH proton RT (PRT) using double scattered protons with computed tomography guidance and provide the first report of proton FLASH RT-mediated normal tissue radioprotection. METHODS AND MATERIALS Absolute dose was measured at multiple depths in solid water and validated against an absolute integral charge measurement using a Faraday cup. Real-time dose rate was obtained using a NaI detector to measure prompt gamma rays. The effect of FLASH versus standard dose rate PRT on tumors and normal tissues was measured using pancreatic flank tumors (MH641905) derived from the KPC autochthonous PanCa model in syngeneic C57BL/6J mice with analysis of fibrosis and stem cell repopulation in small intestine after abdominal irradiation. RESULTS The double scattering and collimation apparatus was dosimetrically validated with dose rates of 78 ± 9 Gy per second and 0.9 ± 0.08 Gy per second for the FLASH and standard PRT. Whole abdominal FLASH PRT at 15 Gy significantly reduced the loss of proliferating cells in intestinal crypts compared with standard PRT. Studies with local intestinal irradiation at 18 Gy revealed a reduction to near baseline levels of intestinal fibrosis for FLASH-PRT compared with standard PRT. Despite this difference, FLASH-PRT did not demonstrate tumor radioprotection in MH641905 pancreatic cancer flank tumors after 12 or 18 Gy irradiation. CONCLUSIONS We have designed and dosimetrically validated a FLASH-PRT system with accurate control of beam flux on a millisecond time scale and online monitoring of the integral and dose delivery time structure. Using this system, we found that FLASH-PRT decreases acute cell loss and late fibrosis after whole-abdomen and focal intestinal RT, whereas tumor growth inhibition is preserved between the 2 modalities.
Collapse
Affiliation(s)
- Eric S Diffenderfer
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ioannis I Verginadis
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michele M Kim
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Khayrullo Shoniyozov
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anastasia Velalopoulou
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Denisa Goia
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mary Putt
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sarah Hagan
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Stephen Avery
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kevin Teo
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Wei Zou
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alexander Lin
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Samuel Swisher-McClure
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Cameron Koch
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ann R Kennedy
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Andy Minn
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Amit Maity
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Theresa M Busch
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lei Dong
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Costas Koumenis
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - James Metz
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Keith A Cengel
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
23
|
Stripay JL, Merchant TE, Roussel MF, Tinkle CL. Preclinical Models of Craniospinal Irradiation for Medulloblastoma. Cancers (Basel) 2020; 12:cancers12010133. [PMID: 31948065 PMCID: PMC7016884 DOI: 10.3390/cancers12010133] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 01/02/2020] [Accepted: 01/02/2020] [Indexed: 02/07/2023] Open
Abstract
Medulloblastoma is an embryonal tumor that shows a predilection for distant metastatic spread and leptomeningeal seeding. For most patients, optimal management of medulloblastoma includes maximum safe resection followed by adjuvant craniospinal irradiation (CSI) and chemotherapy. Although CSI is crucial in treating medulloblastoma, the realization that medulloblastoma is a heterogeneous disease comprising four distinct molecular subgroups (wingless [WNT], sonic hedgehog [SHH], Group 3 [G3], and Group 4 [G4]) with distinct clinical characteristics and prognoses has refocused efforts to better define the optimal role of CSI within and across disease subgroups. The ability to deliver clinically relevant CSI to preclinical models of medulloblastoma offers the potential to study radiation dose and volume effects on tumor control and toxicity in these subgroups and to identify subgroup-specific combination adjuvant therapies. Recent efforts have employed commercial image-guided small animal irradiation systems as well as custom approaches to deliver accurate and reproducible fractionated CSI in various preclinical models of medulloblastoma. Here, we provide an overview of the current clinical indications for, and technical aspects of, irradiation of pediatric medulloblastoma. We then review the current literature on preclinical modeling of and treatment interventions for medulloblastoma and conclude with a summary of challenges in the field of preclinical modeling of CSI for the treatment of leptomeningeal seeding tumors.
Collapse
Affiliation(s)
- Jennifer L. Stripay
- Departments of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (J.L.S.); (M.F.R.)
| | - Thomas E. Merchant
- Departments of Radiation Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Martine F. Roussel
- Departments of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (J.L.S.); (M.F.R.)
| | - Christopher L. Tinkle
- Departments of Radiation Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
- Correspondence: ; Tel.: +1-(901)-595-8735; Fax: +1-(901)-595-3113
| |
Collapse
|