101
|
Yap J, De Franco A, Sheehy S. Future Developments in Charged Particle Therapy: Improving Beam Delivery for Efficiency and Efficacy. Front Oncol 2021; 11:780025. [PMID: 34956897 PMCID: PMC8697351 DOI: 10.3389/fonc.2021.780025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/16/2021] [Indexed: 01/09/2023] Open
Abstract
The physical and clinical benefits of charged particle therapy (CPT) are well recognized. However, the availability of CPT and complete exploitation of dosimetric advantages are still limited by high facility costs and technological challenges. There are extensive ongoing efforts to improve upon these, which will lead to greater accessibility, superior delivery, and therefore better treatment outcomes. Yet, the issue of cost remains a primary hurdle as utility of CPT is largely driven by the affordability, complexity and performance of current technology. Modern delivery techniques are necessary but limited by extended treatment times. Several of these aspects can be addressed by developments in the beam delivery system (BDS) which determines the overall shaping and timing capabilities enabling high quality treatments. The energy layer switching time (ELST) is a limiting constraint of the BDS and a determinant of the beam delivery time (BDT), along with the accelerator and other factors. This review evaluates the delivery process in detail, presenting the limitations and developments for the BDS and related accelerator technology, toward decreasing the BDT. As extended BDT impacts motion and has dosimetric implications for treatment, we discuss avenues to minimize the ELST and overview the clinical benefits and feasibility of a large energy acceptance BDS. These developments support the possibility of advanced modalities and faster delivery for a greater range of treatment indications which could also further reduce costs. Further work to realize methodologies such as volumetric rescanning, FLASH, arc, multi-ion and online image guided therapies are discussed. In this review we examine how increased treatment efficiency and efficacy could be achieved with improvements in beam delivery and how this could lead to faster and higher quality treatments for the future of CPT.
Collapse
Affiliation(s)
- Jacinta Yap
- School of Physics, University of Melbourne, Melbourne, VIC, Australia
| | - Andrea De Franco
- IFMIF Accelerator Development Group, Rokkasho Fusion Institute, National Institutes for Quantum Science and Technology, Aomori, Japan
| | - Suzie Sheehy
- School of Physics, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
102
|
Nachankar A, Oike T, Hanaoka H, Kanai A, Sato H, Yoshida Y, Obinata H, Sakai M, Osu N, Hirota Y, Takahashi A, Shibata A, Ohno T. 64Cu-ATSM Predicts Efficacy of Carbon Ion Radiotherapy Associated with Cellular Antioxidant Capacity. Cancers (Basel) 2021; 13:cancers13246159. [PMID: 34944777 PMCID: PMC8699283 DOI: 10.3390/cancers13246159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/13/2021] [Accepted: 12/03/2021] [Indexed: 12/17/2022] Open
Abstract
Simple Summary Carbon ion radiotherapy is an emerging cancer treatment modality that has a greater therapeutic window than conventional photon radiotherapy. To maximize the efficacy of this extremely scarce medical resource, it is important to identify predictive biomarkers of higher carbon ion relative biological effectiveness (RBE) over photons. Here we show that the carbon ion RBE in human cancer cells correlates with the cellular uptake of 64Cu(II)-diacetyl-bis(N4-methylthiosemicarbazone) (64Cu-ATSM), a potential radioligand that reflects an over-reduced intracellular environment. High RBE/64Cu-ATSM cells show greater steady-state levels of antioxidant proteins and increased capacity to scavenge reactive oxygen species in response to X-rays than low RBE/64Cu-ATSM counterparts. These data suggest that the cellular antioxidant activity is a possible determinant of carbon ion RBE predictable by 64Cu-ATSM uptake. Abstract Carbon ion radiotherapy is an emerging cancer treatment modality that has a greater therapeutic window than conventional photon radiotherapy. To maximize the efficacy of this extremely scarce medical resource, it is important to identify predictive biomarkers of higher carbon ion relative biological effectiveness (RBE) over photons. We addressed this issue by focusing on cellular antioxidant capacity and investigated 64Cu(II)-diacetyl-bis(N4-methylthiosemicarbazone) (64Cu-ATSM), a potential radioligand that reflects an over-reduced intracellular environment. We found that the carbon ion RBE correlated with 64Cu-ATSM uptake both in vitro and in vivo. High RBE/64Cu-ATSM cells showed greater steady-state levels of antioxidant proteins and increased capacity to scavenge reactive oxygen species in response to X-rays than low RBE/64Cu-ATSM counterparts; this upregulation of antioxidant systems was associated with downregulation of TCA cycle intermediates. Furthermore, inhibition of nuclear factor erythroid 2-related factor 2 (Nrf2) sensitized high RBE/64Cu-ATSM cells to X-rays, thereby reducing RBE values to levels comparable to those in low RBE/64Cu-ATSM cells. These data suggest that the cellular activity of Nrf2-driven antioxidant systems is a possible determinant of carbon ion RBE predictable by 64Cu-ATSM uptake. These new findings highlight the potential clinical utility of 64Cu-ATSM imaging to identify high RBE tumors that will benefit from carbon ion radiotherapy.
Collapse
Affiliation(s)
- Ankita Nachankar
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan; (A.N.); (H.S.); (N.O.); (Y.H.); (T.O.)
| | - Takahiro Oike
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan; (A.N.); (H.S.); (N.O.); (Y.H.); (T.O.)
- Gunma University Heavy Ion Medical Center, Maebashi 371-8511, Japan; (Y.Y.); (M.S.); (A.T.)
- Correspondence: ; Tel.: +81-27-220-8383
| | - Hirofumi Hanaoka
- Department of Radiotheranostics, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan; (H.H.); (A.K.)
| | - Ayaka Kanai
- Department of Radiotheranostics, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan; (H.H.); (A.K.)
| | - Hiro Sato
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan; (A.N.); (H.S.); (N.O.); (Y.H.); (T.O.)
- Gunma University Heavy Ion Medical Center, Maebashi 371-8511, Japan; (Y.Y.); (M.S.); (A.T.)
| | - Yukari Yoshida
- Gunma University Heavy Ion Medical Center, Maebashi 371-8511, Japan; (Y.Y.); (M.S.); (A.T.)
| | - Hideru Obinata
- Laboratory for Analytical Instruments, Education and Research Support Center, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan;
| | - Makoto Sakai
- Gunma University Heavy Ion Medical Center, Maebashi 371-8511, Japan; (Y.Y.); (M.S.); (A.T.)
| | - Naoto Osu
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan; (A.N.); (H.S.); (N.O.); (Y.H.); (T.O.)
| | - Yuka Hirota
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan; (A.N.); (H.S.); (N.O.); (Y.H.); (T.O.)
| | - Akihisa Takahashi
- Gunma University Heavy Ion Medical Center, Maebashi 371-8511, Japan; (Y.Y.); (M.S.); (A.T.)
| | - Atsushi Shibata
- Signal Transduction Program, Gunma University Initiative for Advanced Research (GIAR), Maebashi 371-8511, Japan;
| | - Tatsuya Ohno
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Maebashi 371-8511, Japan; (A.N.); (H.S.); (N.O.); (Y.H.); (T.O.)
- Gunma University Heavy Ion Medical Center, Maebashi 371-8511, Japan; (Y.Y.); (M.S.); (A.T.)
| |
Collapse
|
103
|
Orlandi E, Ferrari M, Lafe E, Preda L, Benazzo M, Vischioni B, Bonora M, Rampinelli V, Schreiber A, Licitra L, Nicolai P. When Everything Revolves Around Internal Carotid Artery: Analysis of Different Management Strategies in Patients With Very Advanced Cancer Involving the Skull Base. Front Oncol 2021; 11:781205. [PMID: 34869033 PMCID: PMC8636461 DOI: 10.3389/fonc.2021.781205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/02/2021] [Indexed: 11/13/2022] Open
Abstract
Internal or common carotid artery encasement (CAE) is observed in almost 2-7% of head and neck cancers (HNC) and designates the tumor with the T4b category. This clinical scenario is associated with a dismal prognosis, owing to the risk for thrombosis and bleeding that usually characterizes such an advanced cancer. Standardized radiological criteria to infer invasion of the carotid artery are lacking. Complete surgical resection in the context of a multimodality treatment is supposed to offer the greatest chances of cure. Surgery can either be carotid-sparing or include carotidectomy. Data on probability of cerebrovascular and non-cerebrovascular complications, risk of carotid blowout, poor oncologic outcomes, and less-than-certain efficacy of diagnostic and interventional preventive procedures against cerebral infarction make it difficult to define surgery as the recommended option among other therapeutic strategies. Non-surgical therapies based on radiation therapy possibly combined with chemotherapy are more frequently employed in HNC with CAE. In this context, carotid blowout is the most feared complication, and its probability increases with tumor stage and cumulative radiation dose received by the vessel. The use of highly conformal radiotherapies such as intensity-modulated particle therapy might substantially improve the manageability of HNC with CAE by possibly reducing the risk of late sequalae. Despite evidence is frail, it appears logical that a case-by-case evaluation through multidisciplinary decision making between head and neck surgeons, radiation oncologists, medical oncologists, diagnostic and interventional radiologists, and vascular surgeons are of paramount value to offer the best therapeutic solution to patients affected by HNC with CAE.
Collapse
Affiliation(s)
- Ester Orlandi
- Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy ("Fondazione CNAO"), Pavia, Italy
| | - Marco Ferrari
- Section of Otorhinolaryngology - Head and Neck Surgery, Department of Neurosciences, University of Padua - "Azienda Ospedaliera di Padova", Padua, Italy.,University Health Network (UHN) Guided Therapeutics (GTx) Program International Scholar, UHN, Toronto, ON, Canada.,Technology for Health (PhD Program), Department of Information Engineering, University of Brescia, Brescia, Italy
| | - Elvis Lafe
- Department of Diagnostic Radiology and Interventional Radiology and Neuroradiology, University of Pavia, IRCCS Policlinico San Matteo Foundation, Pavia, Italy.,Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Lorenzo Preda
- Department of Diagnostic Radiology and Interventional Radiology and Neuroradiology, University of Pavia, IRCCS Policlinico San Matteo Foundation, Pavia, Italy.,Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Marco Benazzo
- Department of Otorhinolaryngology, University of Pavia, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Barbara Vischioni
- Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy ("Fondazione CNAO"), Pavia, Italy
| | - Maria Bonora
- Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy ("Fondazione CNAO"), Pavia, Italy
| | - Vittorio Rampinelli
- Technology for Health (PhD Program), Department of Information Engineering, University of Brescia, Brescia, Italy.,Unit of Otorhinolaryngology - Head and Neck Surgery, University of Brescia - "ASST Spedali Civili di Brescia", Brescia, Italy
| | - Alberto Schreiber
- Unit of Otorhinolaryngology - Head and Neck Surgery, University of Brescia - "ASST Spedali Civili di Brescia", Brescia, Italy
| | - Lisa Licitra
- Department of Oncology and Hematology-Oncology, University of Milan, IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Piero Nicolai
- Section of Otorhinolaryngology - Head and Neck Surgery, Department of Neurosciences, University of Padua - "Azienda Ospedaliera di Padova", Padua, Italy
| |
Collapse
|
104
|
Avoidance or adaptation of radiotherapy in patients with cancer with Li-Fraumeni and heritable TP53-related cancer syndromes. Lancet Oncol 2021; 22:e562-e574. [PMID: 34856153 DOI: 10.1016/s1470-2045(21)00425-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/14/2021] [Accepted: 07/21/2021] [Indexed: 12/18/2022]
Abstract
The management of patients with cancer and Li-Fraumeni or heritable TP53-related cancer syndromes is complex because of their increased risk of developing second malignant neoplasms after genotoxic stresses such as systemic treatments or radiotherapy (radiosusceptibility). Clinical decision making also integrates the risks of normal tissue toxicity and sequelae (radiosensitivity) and tumour response to radiotherapy (radioresistance and radiocurability). Radiotherapy should be avoided in patients with cancer and Li-Fraumeni or heritable TP53 cancer-related syndromes, but overall prognosis might be poor without radiotherapy: radioresistance in these patients seems similar to or worse than that of the general population. Radiosensitivity in germline TP53 variant carriers seems similar to that in the general population. The risk of second malignant neoplasms according to germline TP53 variant and the patient's overall oncological prognosis should be assessed during specialised multidisciplinary staff meetings. Radiotherapy should be avoided whenever other similarly curative treatment options are available. In other cases, it should be adapted to minimise the risk of second malignant neoplasms in patients who still require radiotherapy despite its genotoxicity, in view of its potential benefit. Adaptations might be achieved through the reduction of irradiated volumes using proton therapy, non-ionising diagnostic procedures, image guidance, and minimal stray radiation. Non-ionising imaging should become more systematic. Radiotherapy approaches that might result in a lower probability of misrepaired DNA damage (eg, particle therapy biology and tumour targeting) are an area of investigation.
Collapse
|
105
|
Durante M, Debus J, Loeffler JS. Physics and biomedical challenges of cancer therapy with accelerated heavy ions. NATURE REVIEWS. PHYSICS 2021; 3:777-790. [PMID: 34870097 PMCID: PMC7612063 DOI: 10.1038/s42254-021-00368-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/05/2021] [Indexed: 05/05/2023]
Abstract
Radiotherapy should have low toxicity in the entrance channel (normal tissue) and be very effective in cell killing in the target region (tumour). In this regard, ions heavier than protons have both physical and radiobiological advantages over conventional X-rays. Carbon ions represent an excellent combination of physical and biological advantages. There are a dozen carbon-ion clinical centres in Europe and Asia, and more under construction or at the planning stage, including the first in the USA. Clinical results from Japan and Germany are promising, but a heated debate on the cost-effectiveness is ongoing in the clinical community, owing to the larger footprint and greater expense of heavy ion facilities compared with proton therapy centres. We review here the physical basis and the clinical data with carbon ions and the use of different ions, such as helium and oxygen. Research towards smaller and cheaper machines with more effective beam delivery is necessary to make particle therapy affordable. The potential of heavy ions has not been fully exploited in clinics and, rather than there being a single 'silver bullet', different particles and their combination can provide a breakthrough in radiotherapy treatments in specific cases.
Collapse
Affiliation(s)
- Marco Durante
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
- Institute of Condensed Matter Physics, Technische Universität Darmstadt, Darmstadt, Germany
| | - Jürgen Debus
- Department of Radiation Oncology and Heidelberg Ion Beam Therapy Center, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jay S. Loeffler
- Departments of Radiation Oncology and Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
106
|
Huang H, Yuan G, Xu Y, Gao Y, Mao Q, Zhang Y, Bai L, Li W, Wu A, Hu W, Pan Y, Zhou G. Photoacoustic and magnetic resonance imaging-based gene and photothermal therapy using mesoporous nanoagents. Bioact Mater 2021; 9:157-167. [PMID: 34820563 PMCID: PMC8586268 DOI: 10.1016/j.bioactmat.2021.07.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/05/2021] [Accepted: 07/21/2021] [Indexed: 12/20/2022] Open
Abstract
The integration of photothermal therapy (PTT) with gene therapy (GT) in a single nanoscale platform demonstrates great potential in cancer therapy. Porous iron oxide nanoagents (PIONs) are widely used as magnetic nanoagents in the drug delivery field and also serve as a photothermal nanoagent for photothermal therapy. However, the therapeutic efficacy of PIONs-mediated GT has not been studied. The long noncoding RNA (lncRNA) CRYBG3 (LNC CRYBG3), a lncRNA induced by heavy ion irradiation in lung cancer cells, has been reported to directly bind to globular actin (G-actin) and cause degradation of cytoskeleton and blocking of cytokinesis, thus indicating its potential for use in GT by simulating the effect of heavy ion irradiation and functioning as an antitumor drug. In the present study, we investigated the possibility of combining PIONs-mediated PTT and LNC CRYBG3-mediated GT to destroy non-small cell lung cancer (NSCLC) cells both in vitro and in vivo. The combination therapy showed a high cancer cell killing efficacy, and the cure rate was better than that achieved using PTT or GT alone. Moreover, as a type of magnetic nanoagent, PIONs can be used for magnetic resonance imaging (MRI) and photoacoustic imaging (PAI) both in vitro and in vivo. These findings indicate that the new combination therapy has high potential for cancer treatment. LNC CRYBG3 induced by heavy ion irradiation can cause cytoskeleton degradation and function as an antitumor drug. pcDNA3.1-LNC CRYBG3 delivered by PIONs can escape from lysosomes to facilitate plasmid release when exposed to NIR. The combination of PIONs-mediated PTT and LNC CRYBG3-mediated GT presents both diagnosis and treatment potential.
Collapse
Affiliation(s)
- Hao Huang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, PR China
| | - Guotao Yuan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, PR China
| | - Ying Xu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Yuan Gao
- Department of Nutrition and Food Hygiene, Soochow University of Public Health, Suzhou, 215123, China
| | - Qiulian Mao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Yin Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, PR China
| | - Lu Bai
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, PR China
| | - Weijie Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, PR China
| | - Anqing Wu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Wentao Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Yue Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, PR China
| | - Guangming Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| |
Collapse
|
107
|
Ruangchan S, Palmans H, Knäusl B, Georg D, Clausen M. Dose calculation accuracy in particle therapy: Comparing carbon ions with protons. Med Phys 2021; 48:7333-7345. [PMID: 34482555 PMCID: PMC9291072 DOI: 10.1002/mp.15209] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/02/2021] [Accepted: 08/18/2021] [Indexed: 11/18/2022] Open
Abstract
PURPOSE This work presents the validation of an analytical pencil beam dose calculation algorithm in a commercial treatment planning system (TPS) for carbon ions by measurements of dose distributions in heterogeneous phantom geometries. Additionally, a comparison study of carbon ions versus protons is performed considering current best solutions in commercial TPS. METHODS All treatment plans were optimized and calculated using the RayStation TPS (RaySearch, Sweden). The dose distributions calculated with the TPS were compared with measurements using a 24-pinpoint ionization chamber array (T31015, PTW, Germany). Tissue-like inhomogeneities (bone, lung, and soft tissue) were embedded in water, while a target volume of 4 x 4 x 4 cm3 was defined at two different depths behind the heterogeneities. In total, 10 different test cases, with and without range shifter as well as different air gaps, were investigated. Dose distributions inside as well as behind the target volume were evaluated. RESULTS Inside the target volume, the mean dose difference between calculations and measurements, averaged over all test cases, was 1.6% for carbon ions. This compares well to the final agreement of 1.5% obtained in water at the commissioning stage of the TPS for carbon ions and is also within the clinically acceptable interval of 3%. The mean dose difference and maximal dose difference obtained outside the target area were 1.8% and 13.4%, respectively. The agreement of dose distributions for carbon ions in the target volumes was comparable or better to that between Monte Carlo (MC) dose calculations and measurements for protons. Percentage dose differences of more than 10% were present outside the target area behind bone-lung structures, where the carbon ion calculations systematically over predicted the dose. MC dose calculations for protons were superior to carbon ion beams outside the target volumes. CONCLUSION The pencil beam dose calculations for carbon ions in RayStation were found to be in good agreement with dosimetric measurements in heterogeneous geometries for points of interest located within the target. Large local discrepancies behind the target may contribute to incorrect dose predictions for organs at risk.
Collapse
Affiliation(s)
- Sirinya Ruangchan
- Department of Radiation OncologyMedical University of ViennaViennaAustria
- Department of RadiologyKing Chulalongkorn Memorial HospitalBangkokThailand
| | - Hugo Palmans
- Division of Medical PhysicsMedAustron Ion Therapy CenterWiener NeustadtAustria
- Medical Radiation ScienceNational Physical LaboratoryTeddingtonUK
| | - Barbara Knäusl
- Department of Radiation OncologyMedical University of ViennaViennaAustria
- Division of Medical PhysicsMedAustron Ion Therapy CenterWiener NeustadtAustria
| | - Dietmar Georg
- Department of Radiation OncologyMedical University of ViennaViennaAustria
- Division of Medical PhysicsMedAustron Ion Therapy CenterWiener NeustadtAustria
| | - Monika Clausen
- Department of Radiation OncologyMedical University of ViennaViennaAustria
| |
Collapse
|
108
|
Loap P, Vitolo V, Barcellini A, De Marzi L, Mirandola A, Fiore MR, Vischioni B, Jereczek-Fossa BA, Girard N, Kirova Y, Orlandi E. Hadrontherapy for Thymic Epithelial Tumors: Implementation in Clinical Practice. Front Oncol 2021; 11:738320. [PMID: 34707989 PMCID: PMC8543015 DOI: 10.3389/fonc.2021.738320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/21/2021] [Indexed: 12/04/2022] Open
Abstract
Radiation therapy is part of recommendations in the adjuvant settings for advanced stage or as exclusive treatment in unresectable thymic epithelial tumors (TETs). However, first-generation techniques delivered substantial radiation doses to critical organs at risk (OARs), such as the heart or the lungs, resulting in noticeable radiation-induced toxicity. Treatment techniques have significantly evolved for TET irradiation, and modern techniques efficiently spare normal surrounding tissues without negative impact on tumor coverage and consequently local control or patient survival. Considering its dosimetric advantages, hadrontherapy (which includes proton therapy and carbon ion therapy) has proved to be worthwhile for TET irradiation in particular for challenging clinical situations such as cardiac tumoral involvement. However, clinical experience for hadrontherapy is still limited and mainly relies on small-size proton therapy studies. This critical review aims to analyze the current status of hadrontherapy for TET irradiation to implement it at a larger scale.
Collapse
Affiliation(s)
- Pierre Loap
- Department of Radiation Oncology, Institut Curie, Paris, France.,Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Viviana Vitolo
- Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Amelia Barcellini
- Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Ludovic De Marzi
- Department of Radiation Oncology, Institut Curie, Paris, France.,Institut Curie, Paris Sciences & Lettres (PSL) Research University, University Paris Saclay, laboratoire d'Imagerie Translationnelle en Oncologie, Institut National de la Santé et de la Recherche Médicale (INSERM LITO), Orsay, France
| | - Alfredo Mirandola
- Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Maria Rosaria Fiore
- Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Barbara Vischioni
- Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Barbara Alicja Jereczek-Fossa
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy.,Division of Radiotherapy, Istituto Europeo di Oncologia (IEO) European Institute of Oncology Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Nicolas Girard
- Institut du Thorax Curie Montsouris, Paris, France.,Department of Medical Oncology, Institut Curie, Paris, France.,University Paris Saint-Quentin, Versailles, France
| | - Youlia Kirova
- Department of Radiation Oncology, Institut Curie, Paris, France
| | - Ester Orlandi
- Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| |
Collapse
|
109
|
Parisi A, Struelens L, Vanhavere F. Comparison between the results of a recently-developed biological weighting function (V79-RBE 10BWF) and the in vitroclonogenic survival RBE 10of other repair-competent asynchronized normoxic mammalian cell lines and ions not used for the development of the model. Phys Med Biol 2021; 66. [PMID: 34710862 DOI: 10.1088/1361-6560/ac344e] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/28/2021] [Indexed: 11/11/2022]
Abstract
728 simulated microdosimetric lineal energy spectra (26 different ions between 1H and 238U, 28 energy points from 1 to 1000 MeV/n) were used in combination with a recently-developed biological weighting function (Parisi et al., 2020) and 571 published in vitro clonogenic survival curves in order to: 1) assess prediction intervals for the in silico results by deriving an empirical indication of the experimental uncertainty from the dispersion in the in vitro hamster lung fibroblast (V79) data used for the development of the biophysical model; 2) explore the possibility of modeling the relative biological effectiveness (RBE) of the 10% clonogenic survival of asynchronized normoxic repair-competent mammalian cell lines other than the one used for the development of the model (V79); 3) investigate the predictive power of the model through a comparison between in silico results and in vitro data for 10 ions not used for the development of the model. At first, different strategies for the assessment of the in silico prediction intervals were compared. The possible sources of uncertainty responsible for the dispersion in the in vitro data were also shortly reviewed. Secondly, also because of the relevant scatter in the in vitro data, no statistically-relevant differences were found between the RBE10 of the investigated different asynchronized normoxic repair-competent mammalian cell lines. The only exception (Chinese Hamster peritoneal fibroblasts, B14FAF28), is likely due to the limited dataset (all in vitro ion data were extracted from a single publication), systematic differences in the linear energy transfer (LET) calculations for the employed very-heavy ions, and the use of reference photon survival curves extracted from a different publication. Finally, the in silico predictions for the 10 ions not used for the model development were in good agreement with the corresponding in vitro data.
Collapse
Affiliation(s)
- Alessio Parisi
- Radiation Protection Dosimetry and Calibration, Studiecentrum voor Kernenergie, Boeretang 200, Mol, Belgiun, Mol, 2400, BELGIUM
| | - Lara Struelens
- Radiation Protection, Dosimetry and Calibration, Belgian Nuclear Research Centre SCK.CEN, Boeretang 200, Mol, 2400, BELGIUM
| | - Filip Vanhavere
- Institute of Advanced Nuclear Systems, Belgian Nuclear Research Centre SCK.CEN, Boeretang 200, B-2400 Mol, Mol, BELGIUM
| |
Collapse
|
110
|
Barcellini A, Loap P, Murata K, Villa R, Kirova Y, Okonogi N, Orlandi E. PARP Inhibitors in Combination with Radiotherapy: To Do or Not to Do? Cancers (Basel) 2021; 13:cancers13215380. [PMID: 34771545 PMCID: PMC8582502 DOI: 10.3390/cancers13215380] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/18/2021] [Accepted: 10/25/2021] [Indexed: 11/29/2022] Open
Abstract
Simple Summary Despite the large use of inhibitors of Poly-ADP ribose polymerase (PARP-I), the feasibility and safety of their combination with radiotherapy (RT) are unclear. The combination may be particularly interesting in the oligometastatic setting in which patients may benefit from local RT during the treatment with PARP-I. The aim of the current review was to evaluate the outcome and the toxicity in patients with newly diagnosed or recurrent tumors treated with a combination of PARP-I and RT. A total of 12 clinical studies met the inclusion criteria and, despite the heterogeneity of the evaluated patient populations and tumor types, this review suggests that a combination approach is feasible even though the efficacy profile remains unclear. Abstract Background: Despite the large use of inhibitors of Poly-ADP ribose polymerase (PARP-I), the feasibility and safety of their combination with radiotherapy (RT) is unclear. Aim: We conducted a literature analysis with the aim to evaluate the efficacy and safety profile of a combination with RT and PARP-I. Method: The key issues for the current review were expressed in two questions according to the Population, Intervention, Control, Outcome (PICO) criteria: 1. What is the outcome and 2. What is the toxicity in patients treated with a combination of PARP-I and RT for a newly diagnosed or recurrent tumors? Results: A total of 12 clinical studies met the inclusion criteria including seven single-arm dose-escalation phase I studies, two phase II (two- and three-arms controlled trials) trials, one parallel-arm phase I study, and two phase I/II studies published between 2015 and 2021. RT was performed with photon beams and several schedules according to the clinical situation. The acute toxicity ≥ grade 3 ranged between 25% and >96%, which was divided into hematological or non-hematological adverse events. Conclusions: despite the heterogeneity of the evaluated patient populations and tumor types, and the limited number of the studies, this review suggests that a combination approach is feasible even though the efficacy profile remains unclear.
Collapse
Affiliation(s)
- Amelia Barcellini
- Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (A.B.); (R.V.); (E.O.)
| | - Pierre Loap
- Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (A.B.); (R.V.); (E.O.)
- Department of Radiation Oncology, Institut Curie, 75005 Paris, France;
- Correspondence:
| | - Kazutoshi Murata
- National Institutes for Quantum and Radiological Science and Technology, QST Hospital, Chiba 263-0024, Japan; (K.M.); (N.O.)
| | - Riccardo Villa
- Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (A.B.); (R.V.); (E.O.)
| | - Youlia Kirova
- Department of Radiation Oncology, Institut Curie, 75005 Paris, France;
| | - Noriyuki Okonogi
- National Institutes for Quantum and Radiological Science and Technology, QST Hospital, Chiba 263-0024, Japan; (K.M.); (N.O.)
| | - Ester Orlandi
- Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (A.B.); (R.V.); (E.O.)
| |
Collapse
|
111
|
van de Kamp G, Heemskerk T, Kanaar R, Essers J. DNA Double Strand Break Repair Pathways in Response to Different Types of Ionizing Radiation. Front Genet 2021; 12:738230. [PMID: 34659358 PMCID: PMC8514742 DOI: 10.3389/fgene.2021.738230] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/30/2021] [Indexed: 01/12/2023] Open
Abstract
The superior dose distribution of particle radiation compared to photon radiation makes it a promising therapy for the treatment of tumors. However, the cellular responses to particle therapy and especially the DNA damage response (DDR) is not well characterized. Compared to photons, particles are thought to induce more closely spaced DNA lesions instead of isolated lesions. How this different spatial configuration of the DNA damage directs DNA repair pathway usage, is subject of current investigations. In this review, we describe recent insights into induction of DNA damage by particle radiation and how this shapes DNA end processing and subsequent DNA repair mechanisms. Additionally, we give an overview of promising DDR targets to improve particle therapy.
Collapse
Affiliation(s)
- Gerarda van de Kamp
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands.,Oncode Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Tim Heemskerk
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands.,Oncode Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Roland Kanaar
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands.,Oncode Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jeroen Essers
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Vascular Surgery, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Radiation Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
112
|
Riva G, Vischioni B, Gandini S, Cavalieri S, Ronchi S, Barcellini A, Bonora M, Chalaszczyk A, Ingargiola R, Vitolo V, Fiore MR, Iannalfi A, Orlandi E. Particle Beam Therapy Tolerance and Outcome on Patients with Autoimmune Diseases: A Single Institution Matched Case-Control Study. Cancers (Basel) 2021; 13:cancers13205183. [PMID: 34680331 PMCID: PMC8534022 DOI: 10.3390/cancers13205183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 11/16/2022] Open
Abstract
It is unclear whether autoimmune diseases (ADs) may predispose patients to higher radiation-induced toxicity, and no data are available regarding particle therapy. Our objective was to determine if cancer patients with ADs have a higher incidence of complications after protons (PT) or carbon ion (CIRT) therapy. METHODS In our retrospective monocentric study, 38 patients with ADs over 1829 patients were treated with particle therapy between 2011 and 2020. Thirteen patients had collagen vascular disease (CVD), five an inflammatory bowel disease (IBD) and twenty patients an organ-specific AD. Each patient was matched with two control patients without ADs on the basis of type/site of cancer, type of particle treatment, age, sex, hypertension and/or diabetes and previous surgery. RESULTS No G4-5 complications were reported. In the AD group, the frequency of acute grade 3 (G3) toxicity was higher than in the control group (15.8% vs. 2.6%, p = 0.016). Compared to their matched controls, CVD-IBD patients had a higher frequency of G3 acute complications (27.7 vs. 2.6%, p = 0.002). There was no difference between AD patients (7.9%) and controls (2.6%) experiencing late G3 toxicity (p = 0.33). The 2 years disease-free survival was lower in AD patients than in controls (74% vs. 91%, p = 0.01), although the differences in terms of survival were not significant. CONCLUSIONS G3 acute toxicity was more frequently reported in AD patients after PT or CIRT. Since no severe G4-G5 events were reported and in consideration of the benefit of particle therapy for selected cancers, we conclude that particle therapy should be not discouraged for patients with ADs. Further prospective studies are warranted to gain insight into toxicity in cancer patients with ADs enrolled for particle therapy.
Collapse
Affiliation(s)
- Giulia Riva
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (B.V.); (S.R.); (A.B.); (M.B.); (A.C.); (R.I.); (V.V.); (M.R.F.); (A.I.); (E.O.)
- Correspondence: ; Tel.: +39-0382-078-501
| | - Barbara Vischioni
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (B.V.); (S.R.); (A.B.); (M.B.); (A.C.); (R.I.); (V.V.); (M.R.F.); (A.I.); (E.O.)
| | - Sara Gandini
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, 20139 Milan, Italy;
| | - Stefano Cavalieri
- Head and Neck Cancer Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Sara Ronchi
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (B.V.); (S.R.); (A.B.); (M.B.); (A.C.); (R.I.); (V.V.); (M.R.F.); (A.I.); (E.O.)
| | - Amelia Barcellini
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (B.V.); (S.R.); (A.B.); (M.B.); (A.C.); (R.I.); (V.V.); (M.R.F.); (A.I.); (E.O.)
| | - Maria Bonora
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (B.V.); (S.R.); (A.B.); (M.B.); (A.C.); (R.I.); (V.V.); (M.R.F.); (A.I.); (E.O.)
| | - Agnieszka Chalaszczyk
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (B.V.); (S.R.); (A.B.); (M.B.); (A.C.); (R.I.); (V.V.); (M.R.F.); (A.I.); (E.O.)
| | - Rossana Ingargiola
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (B.V.); (S.R.); (A.B.); (M.B.); (A.C.); (R.I.); (V.V.); (M.R.F.); (A.I.); (E.O.)
| | - Viviana Vitolo
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (B.V.); (S.R.); (A.B.); (M.B.); (A.C.); (R.I.); (V.V.); (M.R.F.); (A.I.); (E.O.)
| | - Maria Rosaria Fiore
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (B.V.); (S.R.); (A.B.); (M.B.); (A.C.); (R.I.); (V.V.); (M.R.F.); (A.I.); (E.O.)
| | - Alberto Iannalfi
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (B.V.); (S.R.); (A.B.); (M.B.); (A.C.); (R.I.); (V.V.); (M.R.F.); (A.I.); (E.O.)
| | - Ester Orlandi
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), 27100 Pavia, Italy; (B.V.); (S.R.); (A.B.); (M.B.); (A.C.); (R.I.); (V.V.); (M.R.F.); (A.I.); (E.O.)
| |
Collapse
|
113
|
Averbeck D, Rodriguez-Lafrasse C. Role of Mitochondria in Radiation Responses: Epigenetic, Metabolic, and Signaling Impacts. Int J Mol Sci 2021; 22:ijms222011047. [PMID: 34681703 PMCID: PMC8541263 DOI: 10.3390/ijms222011047] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/24/2021] [Accepted: 10/08/2021] [Indexed: 12/15/2022] Open
Abstract
Until recently, radiation effects have been considered to be mainly due to nuclear DNA damage and their management by repair mechanisms. However, molecular biology studies reveal that the outcomes of exposures to ionizing radiation (IR) highly depend on activation and regulation through other molecular components of organelles that determine cell survival and proliferation capacities. As typical epigenetic-regulated organelles and central power stations of cells, mitochondria play an important pivotal role in those responses. They direct cellular metabolism, energy supply and homeostasis as well as radiation-induced signaling, cell death, and immunological responses. This review is focused on how energy, dose and quality of IR affect mitochondria-dependent epigenetic and functional control at the cellular and tissue level. Low-dose radiation effects on mitochondria appear to be associated with epigenetic and non-targeted effects involved in genomic instability and adaptive responses, whereas high-dose radiation effects (>1 Gy) concern therapeutic effects of radiation and long-term outcomes involving mitochondria-mediated innate and adaptive immune responses. Both effects depend on radiation quality. For example, the increased efficacy of high linear energy transfer particle radiotherapy, e.g., C-ion radiotherapy, relies on the reduction of anastasis, enhanced mitochondria-mediated apoptosis and immunogenic (antitumor) responses.
Collapse
Affiliation(s)
- Dietrich Averbeck
- Laboratory of Cellular and Molecular Radiobiology, PRISME, UMR CNRS 5822/IN2P3, IP2I, Lyon-Sud Medical School, University Lyon 1, 69921 Oullins, France;
- Correspondence:
| | - Claire Rodriguez-Lafrasse
- Laboratory of Cellular and Molecular Radiobiology, PRISME, UMR CNRS 5822/IN2P3, IP2I, Lyon-Sud Medical School, University Lyon 1, 69921 Oullins, France;
- Department of Biochemistry and Molecular Biology, Lyon-Sud Hospital, Hospices Civils de Lyon, 69310 Pierre-Bénite, France
| |
Collapse
|
114
|
Zakaria AM, Colangelo NW, Meesungnoen J, Jay-Gerin JP. Transient hypoxia in water irradiated by swift carbon ions at ultra-high dose rates: implication for FLASH carbon-ion therapy. CAN J CHEM 2021. [DOI: 10.1139/cjc-2021-0110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Large doses of ionizing radiation delivered to tumors at ultra-high dose rates (i.e., in a few milliseconds) paradoxically spare the surrounding healthy tissue while preserving anti-tumor activity (compared with conventional radiotherapy delivered at much lower dose rates). This new modality is known as “FLASH radiotherapy” (FLASH-RT). Although the molecular mechanisms underlying FLASH-RT are not yet fully understood, it has been suggested that radiation delivered at high dose rates spares normal tissue via oxygen depletion followed by subsequent radioresistance of the irradiated tissue. To date, FLASH-RT has been studied using electrons, photons, and protons in various basic biological experiments, pre-clinical studies, and recently in a human patient. However, the efficacy of heavy ions, such as energetic carbon ions, under FLASH-RT conditions remains unclear. Given that living cells and tissues consist mainly of water, we set out to study, from a pure radiation chemistry perspective, the effects of ultra-high dose rates on the transient yields and concentrations of radiolytic species formed in water irradiated by 300-MeV per nucleon carbon ions (LET ∼ 11.6 keV/µm). This mimics irradiation in the “plateau” region of the depth–dose distribution of ions, i.e., in the “normal” tissue region in which the LET is rather low. We used Monte Carlo simulations of multiple, simultaneously interacting radiation tracks together with an “instantaneous pulse” irradiation model. Our calculations show a pronounced oxygen depletion around 0.2 μs, strongly suggesting, as with electrons, photons, and protons, that irradiation with energetic carbon ions at ultra-high dose rates is suitable for FLASH-RT.
Collapse
Affiliation(s)
- Abdullah Muhammad Zakaria
- Département de médecine nucléaire et de radiobiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001, 12ème Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Nicholas W. Colangelo
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, NY, USA
| | - Jintana Meesungnoen
- Département de médecine nucléaire et de radiobiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001, 12ème Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Jean-Paul Jay-Gerin
- Département de médecine nucléaire et de radiobiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001, 12ème Avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| |
Collapse
|
115
|
Therapeutic Modification of Hypoxia. Clin Oncol (R Coll Radiol) 2021; 33:e492-e509. [PMID: 34535359 DOI: 10.1016/j.clon.2021.08.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 08/04/2021] [Accepted: 08/27/2021] [Indexed: 12/30/2022]
Abstract
Regions of reduced oxygenation (hypoxia) are a characteristic feature of virtually all animal and human solid tumours. Numerous preclinical studies, both in vitro and in vivo, have shown that decreasing oxygen concentration induces resistance to radiation. Importantly, hypoxia in human tumours is a negative indicator of radiotherapy outcome. Hypoxia also contributes to resistance to other cancer therapeutics, including immunotherapy, and increases malignant progression as well as cancer cell dissemination. Consequently, substantial effort has been made to detect hypoxia in human tumours and identify realistic approaches to overcome hypoxia and improve cancer therapy outcomes. Hypoxia-targeting strategies include improving oxygen availability, sensitising hypoxic cells to radiation, preferentially killing these cells, locating the hypoxic regions in tumours and increasing the radiation dose to those areas, or applying high energy transfer radiation, which is less affected by hypoxia. Despite numerous clinical studies with each of these hypoxia-modifying approaches, many of which improved both local tumour control and overall survival, hypoxic modification has not been established in routine clinical practice. Here we review the background and significance of hypoxia, how it can be imaged clinically and focus on the various hypoxia-modifying techniques that have undergone, or are currently in, clinical evaluation.
Collapse
|
116
|
Riva G, Imparato S, Savietto G, Pecorilla M, Iannalfi A, Barcellini A, Ronchi S, Fiore MR, Paganelli C, Buizza G, Ciocca M, Baroni G, Preda L, Orlandi E. Potential role of functional imaging in predicting outcome for patients treated with carbon ion therapy: a review. Br J Radiol 2021; 94:20210524. [PMID: 34520670 DOI: 10.1259/bjr.20210524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE Carbon ion radiation therapy (CIRT) is an emerging radiation technique with advantageous physical and radiobiologic properties compared to conventional radiotherapy (RT) providing better response in case of radioresistant and hypoxic tumors. Our aim is to critically review if functional imaging techniques could play a role in predicting outcome of CIRT-treated tumors, as already proven for conventional RT. METHODS 14 studies, concerning Magnetic resonance imaging (MRI) and Positron Emission Tomography (PET), were selected after a comprehensive search on multiple electronic databases from January 2000 to March 2020. RESULTS MRI studies (n = 5) focused on diffusion-weighted MRI and, even though quantitative parameters were the same in all studies (apparent diffusion coefficient, ADC), results were not univocal, probably due to different imaging acquisition protocols and tumoral histology. For PET studies (n = 9), different tracers were used such as [18F]FDG and other uncommon tracers ([11C]MET, [18F]FLT), with a relevant heterogeneity regarding parameters used for outcome assessment. CONCLUSION No conclusion can be drawn on the predictive value of functional imaging in CIRT-treated tumors. A standardization of image acquisition, multi-institutional large trials and external validations are needed in order to establish the prognostic value of functional imaging in CIRT and to guide clinical practice. ADVANCES IN KNOWLEDGE Emerging studies focused on functional imaging's role in predicting CIRT outcome. Due to the heterogeneity of images acquisition and studies, results are conflicting and prospective large studies with imaging standardized protocol are needed.
Collapse
Affiliation(s)
- Giulia Riva
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Sara Imparato
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Giovanni Savietto
- Unit of Radiology, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Mattia Pecorilla
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Alberto Iannalfi
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Amelia Barcellini
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Sara Ronchi
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Maria Rosaria Fiore
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Chiara Paganelli
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, Milano, Italy
| | - Giulia Buizza
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, Milano, Italy
| | - Mario Ciocca
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Guido Baroni
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, Milano, Italy
| | - Lorenzo Preda
- Unit of Radiology, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, Italy.,Department of Radiology, I.R.C.C.S. Policlinico San Matteo Foundation, Pavia, Italy
| | - Ester Orlandi
- Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| |
Collapse
|
117
|
Boscolo D, Kostyleva D, Safari MJ, Anagnostatou V, Äystö J, Bagchi S, Binder T, Dedes G, Dendooven P, Dickel T, Drozd V, Franczack B, Geissel H, Gianoli C, Graeff C, Grahn T, Greiner F, Haettner E, Haghani R, Harakeh MN, Horst F, Hornung C, Hucka JP, Kalantar-Nayestanaki N, Kazantseva E, Kindler B, Knöbel R, Kuzminchuk-Feuerstein N, Lommel B, Mukha I, Nociforo C, Ishikawa S, Lovatti G, Nitta M, Ozoemelam I, Pietri S, Plaß WR, Prochazka A, Purushothaman S, Reidel CA, Roesch H, Schirru F, Schuy C, Sokol O, Steinsberger T, Tanaka YK, Tanihata I, Thirolf P, Tinganelli W, Voss B, Weber U, Weick H, Winfield JS, Winkler M, Zhao J, Scheidenberger C, Parodi K, Durante M. Radioactive Beams for Image-Guided Particle Therapy: The BARB Experiment at GSI. Front Oncol 2021; 11:737050. [PMID: 34504803 PMCID: PMC8422860 DOI: 10.3389/fonc.2021.737050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/04/2021] [Indexed: 12/11/2022] Open
Abstract
Several techniques are under development for image-guidance in particle therapy. Positron (β+) emission tomography (PET) is in use since many years, because accelerated ions generate positron-emitting isotopes by nuclear fragmentation in the human body. In heavy ion therapy, a major part of the PET signals is produced by β+-emitters generated via projectile fragmentation. A much higher intensity for the PET signal can be obtained using β+-radioactive beams directly for treatment. This idea has always been hampered by the low intensity of the secondary beams, produced by fragmentation of the primary, stable beams. With the intensity upgrade of the SIS-18 synchrotron and the isotopic separation with the fragment separator FRS in the FAIR-phase-0 in Darmstadt, it is now possible to reach radioactive ion beams with sufficient intensity to treat a tumor in small animals. This was the motivation of the BARB (Biomedical Applications of Radioactive ion Beams) experiment that is ongoing at GSI in Darmstadt. This paper will present the plans and instruments developed by the BARB collaboration for testing the use of radioactive beams in cancer therapy.
Collapse
Affiliation(s)
- Daria Boscolo
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Daria Kostyleva
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | | | | | - Juha Äystö
- University of Jyväskylä, Jyväskylä, Finland.,Helsinki Institute of Physics, Helsinki, Finland
| | | | - Tim Binder
- Ludwig-Maximilians-Universität München, Munich, Germany
| | | | | | - Timo Dickel
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany.,Justus-Liebig-Universität Gießen, Gießen, Germany
| | - Vasyl Drozd
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany.,University of Groningen, Groningen, Netherlands
| | | | - Hans Geissel
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany.,Justus-Liebig-Universität Gießen, Gießen, Germany
| | | | - Christian Graeff
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Tuomas Grahn
- University of Jyväskylä, Jyväskylä, Finland.,Helsinki Institute of Physics, Helsinki, Finland
| | - Florian Greiner
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Emma Haettner
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | | | | | - Felix Horst
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Christine Hornung
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany.,Technische Universität Darmstadt, Darmstadt, Germany
| | - Jan-Paul Hucka
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany.,Technische Universität Darmstadt, Darmstadt, Germany
| | | | - Erika Kazantseva
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Birgit Kindler
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Ronja Knöbel
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | | | - Bettina Lommel
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Ivan Mukha
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Chiara Nociforo
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | | | | | | | | | - Stephane Pietri
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Wolfgang R Plaß
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany.,Justus-Liebig-Universität Gießen, Gießen, Germany
| | | | | | | | - Heidi Roesch
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany.,Technische Universität Darmstadt, Darmstadt, Germany
| | - Fabio Schirru
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Christoph Schuy
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Olga Sokol
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Timo Steinsberger
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany.,Technische Universität Darmstadt, Darmstadt, Germany
| | | | - Isao Tanihata
- Research Center for Nuclear Physics, Osaka University, Osaka, Japan.,Peking University, Beijing, China.,Institute of Modern Physics, Lanzhou, China
| | - Peter Thirolf
- Ludwig-Maximilians-Universität München, Munich, Germany
| | | | - Bernd Voss
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Uli Weber
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Helmut Weick
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - John S Winfield
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Martin Winkler
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Jianwei Zhao
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany.,Peking University, Beijing, China
| | - Christoph Scheidenberger
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany.,Justus-Liebig-Universität Gießen, Gießen, Germany
| | - Katia Parodi
- Ludwig-Maximilians-Universität München, Munich, Germany
| | - Marco Durante
- GSI Helmholtzzentrum für Schwerionenforschung, Darmstadt, Germany.,Technische Universität Darmstadt, Darmstadt, Germany
| | | |
Collapse
|
118
|
Cavalieri S, Ronchi S, Barcellini A, Bonora M, Vischioni B, Vitolo V, Villa R, Del Vecchio M, Licitra L, Orlandi E. Toxicity of carbon ion radiotherapy and immune checkpoint inhibitors in advanced melanoma. Radiother Oncol 2021; 164:1-5. [PMID: 34506831 DOI: 10.1016/j.radonc.2021.08.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/13/2021] [Accepted: 08/28/2021] [Indexed: 01/04/2023]
Abstract
We analyzed CTCAE adverse events of sequential Carbon Ion radiotherapy (CIRT) and immune checkpoint inhibitors (ICIs) in advanced melanoma patients. The frequencies of early and late adverse events (AEs) were 100% and 82% of patients, respectively. The frequency of G3+ AEs was in line with the literature.
Collapse
Affiliation(s)
- Stefano Cavalieri
- Head and Neck Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sara Ronchi
- Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy.
| | - Amelia Barcellini
- Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Maria Bonora
- Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Barbara Vischioni
- Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Viviana Vitolo
- Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Riccardo Villa
- Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Michele Del Vecchio
- Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Lisa Licitra
- Head and Neck Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Italy
| | - Ester Orlandi
- Radiation Oncology Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| |
Collapse
|
119
|
Particle Radiotherapy for Skull Base Chondrosarcoma: A Clinical Series from Italian National Center for Oncological Hadrontherapy. Cancers (Basel) 2021; 13:cancers13174423. [PMID: 34503233 PMCID: PMC8430859 DOI: 10.3390/cancers13174423] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/09/2021] [Accepted: 08/27/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Skull-base chondrosarcoma is a rare locally aggressive or malignant group of cartilaginous neoplasm. The standard of care consists of surgery and high-dose radiation therapy, better if with particle, due to their radioresistance and proximity to organs at risk such as brainstem and optic pathways. Due to the rarity of the tumor and its site, outcomes in terms of local control and toxicity of patients with this malignancy after receiving particle therapy has been documented only in a limited number of series with a restricted number of patients, in particular with regard to carbon ions. The aim of our retrospective study is to assess the role of particle therapy (protons and carbon ions) after surgery in our Institute in skull-base chondrosarcomas. Abstract Background: The standard treatment for skull base chondrosarcoma (SB-CHS) consists of surgery and high-dose radiation therapy. Our aim was to evaluate outcome in terms of local control (LC) and toxicity of proton therapy (PT) and carbon ion (CIRT) after surgery. Materials and methods: From September 2011 to July 2020, 48 patients underwent particle therapy (67% PT, 33% CIRT) for SB-CHS. PT and CIRT total dose was 70 GyRBE (relative biological effectiveness) in 35 fractions and 70.4 GyRBE in 16 fractions, respectively. Toxicity was assessed using the Common Terminology Criteria for Adverse Events (CTCAE v5). Results: After a median follow-up time of 38 months, one local failure (2%) was documented and the patient died for progressive disease. Overall, 3-year LC was 98%. One (2%) and 4 (8%) patients experienced G3 acute and late toxicity, respectively. White-matter brain changes were documented in 22 (46%) patients, but only 7 needed steroids (G2). No patients had G3 brain toxicity. No G4–5 complications were reported. We did not find any correlation between high-grade toxicity or white-matter changes and characteristics of patients, disease and surgery. Conclusions: PT and CIRT appeared to be effective and safe treatments for patients with SB-CHS, resulting in high LC rates and an acceptable toxicity profile.
Collapse
|
120
|
Cunningham C, de Kock M, Engelbrecht M, Miles X, Slabbert J, Vandevoorde C. Radiosensitization Effect of Gold Nanoparticles in Proton Therapy. Front Public Health 2021; 9:699822. [PMID: 34395371 PMCID: PMC8358148 DOI: 10.3389/fpubh.2021.699822] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 06/30/2021] [Indexed: 01/02/2023] Open
Abstract
The number of proton therapy facilities and the clinical usage of high energy proton beams for cancer treatment has substantially increased over the last decade. This is mainly due to the superior dose distribution of proton beams resulting in a reduction of side effects and a lower integral dose compared to conventional X-ray radiotherapy. More recently, the usage of metallic nanoparticles as radiosensitizers to enhance radiotherapy is receiving growing attention. While this strategy was originally intended for X-ray radiotherapy, there is currently a small number of experimental studies indicating promising results for proton therapy. However, most of these studies used low proton energies, which are less applicable to clinical practice; and very small gold nanoparticles (AuNPs). Therefore, this proof of principle study evaluates the radiosensitization effect of larger AuNPs in combination with a 200 MeV proton beam. CHO-K1 cells were exposed to a concentration of 10 μg/ml of 50 nm AuNPs for 4 hours before irradiation with a clinical proton beam at NRF iThemba LABS. AuNP internalization was confirmed by inductively coupled mass spectrometry and transmission electron microscopy, showing a random distribution of AuNPs throughout the cytoplasm of the cells and even some close localization to the nuclear membrane. The combined exposure to AuNPs and protons resulted in an increase in cell killing, which was 27.1% at 2 Gy and 43.8% at 6 Gy, compared to proton irradiation alone, illustrating the radiosensitizing potential of AuNPs. Additionally, cells were irradiated at different positions along the proton depth-dose curve to investigate the LET-dependence of AuNP radiosensitization. An increase in cytogenetic damage was observed at all depths for the combined treatment compared to protons alone, but no incremental increase with LET could be determined. In conclusion, this study confirms the potential of 50 nm AuNPs to increase the therapeutic efficacy of proton therapy.
Collapse
Affiliation(s)
- Charnay Cunningham
- Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, National Research Foundation, Cape Town, South Africa.,Department of Medical Biosciences, Faculty of Natural Sciences, University of the Western Cape, Cape Town, South Africa
| | - Maryna de Kock
- Department of Medical Biosciences, Faculty of Natural Sciences, University of the Western Cape, Cape Town, South Africa
| | - Monique Engelbrecht
- Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, National Research Foundation, Cape Town, South Africa.,Department of Medical Biosciences, Faculty of Natural Sciences, University of the Western Cape, Cape Town, South Africa
| | - Xanthene Miles
- Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, National Research Foundation, Cape Town, South Africa
| | - Jacobus Slabbert
- Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, National Research Foundation, Cape Town, South Africa
| | - Charlot Vandevoorde
- Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, National Research Foundation, Cape Town, South Africa
| |
Collapse
|
121
|
Weber UA, Scifoni E, Durante M. FLASH radiotherapy with carbon ion beams. Med Phys 2021; 49:1974-1992. [PMID: 34318508 DOI: 10.1002/mp.15135] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/19/2021] [Accepted: 07/19/2021] [Indexed: 12/17/2022] Open
Abstract
FLASH radiotherapy is considered a new potential breakthrough in cancer treatment. Ultra-high dose rates (>40 Gy/s) have been shown to reduce toxicity in the normal tissue without compromising tumor control, resulting in a widened therapeutic window. These high dose rates are more easily achievable in the clinic with charged particles, and clinical trials are, indeed, ongoing using electrons or protons. FLASH could be an attractive solution also for heavier ions such as carbon and could even enhance the therapeutic window. However, it is not yet known whether the FLASH effect will be the same as for sparsely ionizing radiation when densely ionizing carbons ions are used. Here we discuss the technical challenges in beam delivery and present a promising solution using 3D range-modulators in order to apply ultra-high dose rates (UHDR) compatible with FLASH with carbon ions. Furthermore, we will discuss the possible outcome of C-ion therapy at UHDR on the level of the radiobiological and radiation chemical effects.
Collapse
Affiliation(s)
- Uli Andreas Weber
- Biophysics Department, GSI Helhmoltzzentrum für Schwerionenforschung, Darmstadt, Germany
| | - Emanuele Scifoni
- Istituto Nazionale di Fisica Nucleare (INFN), Trento Institute for Fundamental Physics and Applications (TIFPA), Trento, Italy
| | - Marco Durante
- Biophysics Department, GSI Helhmoltzzentrum für Schwerionenforschung, Darmstadt, Germany.,Institute of Condensed Matter Physics, Technische Universität Darmstadt, Darmstadt, Germany
| |
Collapse
|
122
|
Lorat Y, Reindl J, Isermann A, Rübe C, Friedl AA, Rübe CE. Focused Ion Microbeam Irradiation Induces Clustering of DNA Double-Strand Breaks in Heterochromatin Visualized by Nanoscale-Resolution Electron Microscopy. Int J Mol Sci 2021; 22:ijms22147638. [PMID: 34299263 PMCID: PMC8306362 DOI: 10.3390/ijms22147638] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/06/2021] [Accepted: 07/15/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Charged-particle radiotherapy is an emerging treatment modality for radioresistant tumors. The enhanced effectiveness of high-energy particles (such as heavy ions) has been related to the spatial clustering of DNA lesions due to highly localized energy deposition. Here, DNA damage patterns induced by single and multiple carbon ions were analyzed in the nuclear chromatin environment by different high-resolution microscopy approaches. Material and Methods: Using the heavy-ion microbeam SNAKE, fibroblast monolayers were irradiated with defined numbers of carbon ions (1/10/100 ions per pulse, ipp) focused to micrometer-sized stripes or spots. Radiation-induced lesions were visualized as DNA damage foci (γH2AX, 53BP1) by conventional fluorescence and stimulated emission depletion (STED) microscopy. At micro- and nanoscale level, DNA double-strand breaks (DSBs) were visualized within their chromatin context by labeling the Ku heterodimer. Single and clustered pKu70-labeled DSBs were quantified in euchromatic and heterochromatic regions at 0.1 h, 5 h and 24 h post-IR by transmission electron microscopy (TEM). Results: Increasing numbers of carbon ions per beam spot enhanced spatial clustering of DNA lesions and increased damage complexity with two or more DSBs in close proximity. This effect was detectable in euchromatin, but was much more pronounced in heterochromatin. Analyzing the dynamics of damage processing, our findings indicate that euchromatic DSBs were processed efficiently and repaired in a timely manner. In heterochromatin, by contrast, the number of clustered DSBs continuously increased further over the first hours following IR exposure, indicating the challenging task for the cell to process highly clustered DSBs appropriately. Conclusion: Increasing numbers of carbon ions applied to sub-nuclear chromatin regions enhanced the spatial clustering of DSBs and increased damage complexity, this being more pronounced in heterochromatic regions. Inefficient processing of clustered DSBs may explain the enhanced therapeutic efficacy of particle-based radiotherapy in cancer treatment.
Collapse
Affiliation(s)
- Yvonne Lorat
- Department of Radiation Oncology, Saarland University Hospital, 66421 Homburg, Germany; (Y.L.); (A.I.); (C.R.)
| | - Judith Reindl
- Institute for Applied Physic and Metrology, Universität der Bundeswehr München, 85577 Neubiberg, Germany;
| | - Anna Isermann
- Department of Radiation Oncology, Saarland University Hospital, 66421 Homburg, Germany; (Y.L.); (A.I.); (C.R.)
| | - Christian Rübe
- Department of Radiation Oncology, Saarland University Hospital, 66421 Homburg, Germany; (Y.L.); (A.I.); (C.R.)
| | - Anna A. Friedl
- Department of Radiation Oncology, University Hospital, Ludwig-Maximilian University, 80539 Munich, Germany;
| | - Claudia E. Rübe
- Department of Radiation Oncology, Saarland University Hospital, 66421 Homburg, Germany; (Y.L.); (A.I.); (C.R.)
- Correspondence: ; Tel.: +49-6841-1634614
| |
Collapse
|
123
|
Miles X, Vandevoorde C, Hunter A, Bolcaen J. MDM2/X Inhibitors as Radiosensitizers for Glioblastoma Targeted Therapy. Front Oncol 2021; 11:703442. [PMID: 34307171 PMCID: PMC8296304 DOI: 10.3389/fonc.2021.703442] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/24/2021] [Indexed: 12/24/2022] Open
Abstract
Inhibition of the MDM2/X-p53 interaction is recognized as a potential anti-cancer strategy, including the treatment of glioblastoma (GB). In response to cellular stressors, such as DNA damage, the tumor suppression protein p53 is activated and responds by mediating cellular damage through DNA repair, cell cycle arrest and apoptosis. Hence, p53 activation plays a central role in cell survival and the effectiveness of cancer therapies. Alterations and reduced activity of p53 occur in 25-30% of primary GB tumors, but this number increases drastically to 60-70% in secondary GB. As a result, reactivating p53 is suggested as a treatment strategy, either by using targeted molecules to convert the mutant p53 back to its wild type form or by using MDM2 and MDMX (also known as MDM4) inhibitors. MDM2 down regulates p53 activity via ubiquitin-dependent degradation and is amplified or overexpressed in 14% of GB cases. Thus, suppression of MDM2 offers an opportunity for urgently needed new therapeutic interventions for GB. Numerous small molecule MDM2 inhibitors are currently undergoing clinical evaluation, either as monotherapy or in combination with chemotherapy and/or other targeted agents. In addition, considering the major role of both p53 and MDM2 in the downstream signaling response to radiation-induced DNA damage, the combination of MDM2 inhibitors with radiation may offer a valuable therapeutic radiosensitizing approach for GB therapy. This review covers the role of MDM2/X in cancer and more specifically in GB, followed by the rationale for the potential radiosensitizing effect of MDM2 inhibition. Finally, the current status of MDM2/X inhibition and p53 activation for the treatment of GB is given.
Collapse
Affiliation(s)
- Xanthene Miles
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town, South Africa
| | - Charlot Vandevoorde
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town, South Africa
| | - Alistair Hunter
- Radiobiology Section, Division of Radiation Oncology, Department of Radiation Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
| | - Julie Bolcaen
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town, South Africa
| |
Collapse
|
124
|
Byun HK, Han MC, Yang K, Kim JS, Yoo GS, Koom WS, Kim YB. Physical and Biological Characteristics of Particle Therapy for Oncologists. Cancer Res Treat 2021; 53:611-620. [PMID: 34139805 PMCID: PMC8291193 DOI: 10.4143/crt.2021.066] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022] Open
Abstract
Particle therapy is a promising and evolving modality of radiotherapy that can be used to treat tumors that are radioresistant to conventional photon beam radiotherapy. It has unique biological and physical advantages compared with conventional radiotherapy. The characteristic feature of particle therapy is the "Bragg peak," a steep and localized peak of dose, that enables precise delivery of the radiation dose to the tumor while effectively sparing normal organs. Especially, the charged particles (e.g., proton, helium, carbon) cause a high rate of energy loss along the track, thereby leading to high biological effectiveness, which makes particle therapy attractive. Using this property, the particle beam induces more severe DNA double-strand breaks than the photon beam, which is less influenced by the oxygen level. This review describes the general biological and physical aspects of particle therapy for oncologists, including non-radiation oncologists and beginners in the field.
Collapse
Affiliation(s)
- Hwa Kyung Byun
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Min Cheol Han
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Kyungmi Yang
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jin Sung Kim
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Gyu Sang Yoo
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Woong Sub Koom
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Yong Bae Kim
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
125
|
Durante M. Failla Memorial Lecture: The Many Facets of Heavy-Ion Science. Radiat Res 2021; 195:403-411. [PMID: 33979440 DOI: 10.1667/rade-21-00029.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 02/22/2021] [Indexed: 11/03/2022]
Abstract
Heavy ions are riveting in radiation biophysics, particularly in the areas of radiotherapy and space radiation protection. Accelerated charged particles can indeed penetrate deeply in the human body to sterilize tumors, exploiting the favorable depth-dose distribution of ions compared to conventional X rays. Conversely, the high biological effectiveness in inducing late effects presents a hazard for manned space exploration. Even after half a century of accelerator-based experiments, clinical applications and flight research, these two topics remain both fascinating and baffling. Heavy-ion therapy is very expensive, and despite the clinical success it remains controversial. Research on late radiation morbidity in spaceflight led to a reduction in uncertainty, but also pointed to new risks previously underestimated, such as possible damage to the central nervous system. Recently, heavy ions have also been used in other, unanticipated biomedical fields, such as treatment of heart arrhythmia or inactivation of viruses for vaccine development. Heavy-ion science nicely merges physics and biology and remains an extraordinary research field for the 21st century.
Collapse
Affiliation(s)
- Marco Durante
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany; and Technische Universität Darmstadt, Institute of Condensed Matter Physics, 64289 Darmstadt, Germany
| |
Collapse
|
126
|
Therapeutic challenges in radiation-induced salivary gland cancers. Curr Opin Otolaryngol Head Neck Surg 2021; 29:120-125. [PMID: 33394737 DOI: 10.1097/moo.0000000000000694] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW To give an overview of recent advances in therapeutic approaches of radiation-induced salivary gland cancers (ri-SGCs). RECENT FINDINGS Reirradiation with protons and carbon ions demonstrated to be feasible, safe and to offer good local control rates, with the possibility of overcoming radioresistance and dosimetric issues in previously irradiated cancer patients. Chromosomal rearrangements, gene fusions and expression profiles are important to identify specific cancer subtypes and can guide tailored systemic therapy. SUMMARY Ri-SGCs are rare and heterogeneous. Patients are often heavily pretreated and at risk of toxicities, and their management remain challenging. A multidisciplinary approach in referral centers is mandatory. Knowledge about SGCs cellular and molecular mechanisms is constantly evolving. In the last years, novel advances in therapeutic approaches, such as carbon ion radiotherapy, are emerging as safe and effective options in active treatment, but further efforts are needed to offer tailored personalized treatments and to improve survival.
Collapse
|
127
|
Marcus D, Lieverse RIY, Klein C, Abdollahi A, Lambin P, Dubois LJ, Yaromina A. Charged Particle and Conventional Radiotherapy: Current Implications as Partner for Immunotherapy. Cancers (Basel) 2021; 13:1468. [PMID: 33806808 PMCID: PMC8005048 DOI: 10.3390/cancers13061468] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 02/07/2023] Open
Abstract
Radiotherapy (RT) has been shown to interfere with inflammatory signals and to enhance tumor immunogenicity via, e.g., immunogenic cell death, thereby potentially augmenting the therapeutic efficacy of immunotherapy. Conventional RT consists predominantly of high energy photon beams. Hypofractionated RT regimens administered, e.g., by stereotactic body radiation therapy (SBRT), are increasingly investigated in combination with cancer immunotherapy within clinical trials. Despite intensive preclinical studies, the optimal dose per fraction and dose schemes for elaboration of RT induced immunogenic potential remain inconclusive. Compared to the scenario of combined immune checkpoint inhibition (ICI) and RT, multimodal therapies utilizing other immunotherapy principles such as adoptive transfer of immune cells, vaccination strategies, targeted immune-cytokines and agonists are underrepresented in both preclinical and clinical settings. Despite the clinical success of ICI and RT combination, e.g., prolonging overall survival in locally advanced lung cancer, curative outcomes are still not achieved for most cancer entities studied. Charged particle RT (PRT) has gained interest as it may enhance tumor immunogenicity compared to conventional RT due to its unique biological and physical properties. However, whether PRT in combination with immune therapy will elicit superior antitumor effects both locally and systemically needs to be further investigated. In this review, the immunological effects of RT in the tumor microenvironment are summarized to understand their implications for immunotherapy combinations. Attention will be given to the various immunotherapeutic interventions that have been co-administered with RT so far. Furthermore, the theoretical basis and first evidences supporting a favorable immunogenicity profile of PRT will be examined.
Collapse
Affiliation(s)
- Damiënne Marcus
- The M-Lab, Department of Precision Medicine, GROW–School for Oncology and Developmental Biology, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands; (D.M.); (R.I.Y.L.); (P.L.); (L.J.D.)
| | - Relinde I. Y. Lieverse
- The M-Lab, Department of Precision Medicine, GROW–School for Oncology and Developmental Biology, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands; (D.M.); (R.I.Y.L.); (P.L.); (L.J.D.)
| | - Carmen Klein
- German Cancer Consortium (DKTK) Core-Center Heidelberg, National Center for Tumor Diseases (NCT), Clinical Cooperation Unit Translational Radiation Oncology, Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460, 69120 Heidelberg, Germany; (C.K.); (A.A.)
- Heidelberg Ion-Beam Therapy Center (HIT), Division of Molecular and Translational Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Im Neuenheimer Feld 450, 69120 Heidelberg, Germany
- National Center for Radiation Oncology (NCRO), Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 222, 69120 Heidelberg, Germany
| | - Amir Abdollahi
- German Cancer Consortium (DKTK) Core-Center Heidelberg, National Center for Tumor Diseases (NCT), Clinical Cooperation Unit Translational Radiation Oncology, Heidelberg University Hospital (UKHD) and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460, 69120 Heidelberg, Germany; (C.K.); (A.A.)
- Heidelberg Ion-Beam Therapy Center (HIT), Division of Molecular and Translational Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Im Neuenheimer Feld 450, 69120 Heidelberg, Germany
- National Center for Radiation Oncology (NCRO), Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg University and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 222, 69120 Heidelberg, Germany
| | - Philippe Lambin
- The M-Lab, Department of Precision Medicine, GROW–School for Oncology and Developmental Biology, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands; (D.M.); (R.I.Y.L.); (P.L.); (L.J.D.)
| | - Ludwig J. Dubois
- The M-Lab, Department of Precision Medicine, GROW–School for Oncology and Developmental Biology, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands; (D.M.); (R.I.Y.L.); (P.L.); (L.J.D.)
| | - Ala Yaromina
- The M-Lab, Department of Precision Medicine, GROW–School for Oncology and Developmental Biology, Maastricht University, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands; (D.M.); (R.I.Y.L.); (P.L.); (L.J.D.)
| |
Collapse
|
128
|
Ferrari B, Roda E, Priori EC, De Luca F, Facoetti A, Ravera M, Brandalise F, Locatelli CA, Rossi P, Bottone MG. A New Platinum-Based Prodrug Candidate for Chemotherapy and Its Synergistic Effect With Hadrontherapy: Novel Strategy to Treat Glioblastoma. Front Neurosci 2021; 15:589906. [PMID: 33828444 PMCID: PMC8019820 DOI: 10.3389/fnins.2021.589906] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 02/08/2021] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma (GBM) is the most common tumor of the central nervous system. Current therapies, often associated with severe side effects, are inefficacious to contrast the GBM relapsing forms. In trying to overcome these drawbacks, (OC-6-44)-acetatodiamminedichlorido(2-(2-propynyl)octanoato)platinum(IV), also called Pt(IV)Ac-POA, has been recently synthesized. This new prodrug bearing as axial ligand (2-propynyl)octanoic acid (POA), a histone deacetylase inhibitor, has a higher activity due to (i) its high cellular accumulation by virtue of its high lipophilicity and (ii) the inhibition of histone deacetylase, which leads to the increased exposure of nuclear DNA, permitting higher platination and promoting cancer cell death. In the present study, we investigated the effects induced by Pt(IV)Ac-POA and its potential antitumor activity in human U251 glioblastoma cell line using a battery of complementary techniques, i.e., flow cytometry, immunocytochemistry, TEM, and Western blotting analyses. In addition, the synergistic effect of Pt(IV)Ac-POA associated with the innovative oncological hadrontherapy with carbon ions was investigated, with the aim to identify the most efficient anticancer treatment combination. Our in vitro data demonstrated that Pt(IV)Ac-POA is able to induce cell death, through different pathways, at concentrations lower than those tested for other platinum analogs. In particular, an enduring Pt(IV)Ac-POA antitumor effect, persisting in long-term treatment, was demonstrated. Interestingly, this effect was further amplified by the combined exposure to carbon ion radiation. In conclusion, Pt(IV)Ac-POA represents a promising prodrug to be incorporated into the treatment regimen for GBM. Moreover, the synergistic efficacy of the combined protocol using chemotherapeutic Pt(IV)Ac-POA followed by carbon ion radiation may represent a promising approach, which may overcome some typical limitations of conventional therapeutic protocols for GBM treatment.
Collapse
Affiliation(s)
- Beatrice Ferrari
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Elisa Roda
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy.,Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Erica Cecilia Priori
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Fabrizio De Luca
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Angelica Facoetti
- National Center of Oncological Hadrontherapy (Fondazione CNAO), Pavia, Italy
| | - Mauro Ravera
- Department of Sciences and Technological Innovation (DiSIT), University of Piemonte Orientale "A. Avogadro", Alessandria, Italy
| | - Federico Brandalise
- Department of Fundamental Neurosciences (NEUFO), University of Geneva, Geneva, Switzerland
| | - Carlo Alessandro Locatelli
- Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Paola Rossi
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Maria Grazia Bottone
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| |
Collapse
|
129
|
Prezado Y, Hirayama R, Matsufuji N, Inaniwa T, Martínez-Rovira I, Seksek O, Bertho A, Koike S, Labiod D, Pouzoulet F, Polledo L, Warfving N, Liens A, Bergs J, Shimokawa T. A Potential Renewed Use of Very Heavy Ions for Therapy: Neon Minibeam Radiation Therapy. Cancers (Basel) 2021; 13:cancers13061356. [PMID: 33802835 PMCID: PMC8002595 DOI: 10.3390/cancers13061356] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 01/13/2023] Open
Abstract
Simple Summary The treatment of hypoxic tumours continues to be one of the main challenges for radiation therapy. Minibeam radiation therapy (MBRT) shows a highly promising reduction of to-xicity in normal tissue, so that very heavy ions, such as Neon (Ne) or Argon (Ar), with extremely high LET, might become applicable to clinical situations. The high LET in the target would be unrivalled to overcome hypoxia, while MBRT might limit the side effects normally preventing the use of those heavy ions in a conventional radiotherapeutic setting. The work reported in this manuscript is the first experimental proof of the remarkable reduction of normal tissue (skin) toxicities after Ne MBRT irradiations as compared to conventional Ne irradiations. This result might allow for a renewed use of very heavy ions for cancer therapy. Abstract (1) Background: among all types of radiation, very heavy ions, such as Neon (Ne) or Argon (Ar), are the optimum candidates for hypoxic tumor treatments due to their reduced oxygen enhancement effect. However, their pioneering clinical use in the 1970s was halted due to severe side effects. The aim of this work was to provide a first proof that the combination of very heavy ions with minibeam radiation therapy leads to a minimization of toxicities and, thus, opening the door for a renewed use of heavy ions for therapy; (2) Methods: mouse legs were irradiated with either Ne MBRT or Ne broad beams at the same average dose. Skin toxicity was scored for a period of four weeks. Histopathology evaluations were carried out at the end of the study; (3) Results: a significant difference in toxicity was observed between the two irradiated groups. While severe da-mage, including necrosis, was observed in the broad beam group, only light to mild erythema was present in the MBRT group; (4) Conclusion: Ne MBRT is significantly better tolerated than conventional broad beam irradiations.
Collapse
Affiliation(s)
- Yolanda Prezado
- Institut Curie, Université PSL, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France;
- Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France
- Correspondence:
| | - Ryochi Hirayama
- Department of Charged Particle Therapy Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan; (R.H.); (N.M.); (T.I.); (S.K.); (T.S.)
| | - Naruhiro Matsufuji
- Department of Charged Particle Therapy Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan; (R.H.); (N.M.); (T.I.); (S.K.); (T.S.)
- Department of Accelerator and Medical Physics, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Taku Inaniwa
- Department of Charged Particle Therapy Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan; (R.H.); (N.M.); (T.I.); (S.K.); (T.S.)
- Department of Accelerator and Medical Physics, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Immaculada Martínez-Rovira
- Ionizing Radiation Research Group, Physics Department, Universitat Autònoma de Barcelona (UAB), E-08193 Cerdanyola del Vallès, Spain;
| | - Olivier Seksek
- Université Paris-Saclay, CNRS/IN2P3, Université de Paris, IJCLab, Pole Santé, 91405 Orsay, France;
| | - Annaïg Bertho
- Institut Curie, Université PSL, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France;
- Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France
| | - Sachiko Koike
- Department of Charged Particle Therapy Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan; (R.H.); (N.M.); (T.I.); (S.K.); (T.S.)
- Department of Accelerator and Medical Physics, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Dalila Labiod
- Experimental Radiotherapy Platform, Translational Research Department, Institut Curie, Université Paris Saclay, 91400 Orsay, France; (D.L.); (F.P.)
| | - Frederic Pouzoulet
- Experimental Radiotherapy Platform, Translational Research Department, Institut Curie, Université Paris Saclay, 91400 Orsay, France; (D.L.); (F.P.)
| | - Laura Polledo
- AnaPath Services GmbH, Hammerstrasse 49, 4410 Liestal, Switzerland; (L.P.); (N.W.); (A.L.)
| | - Nils Warfving
- AnaPath Services GmbH, Hammerstrasse 49, 4410 Liestal, Switzerland; (L.P.); (N.W.); (A.L.)
| | - Aléthéa Liens
- AnaPath Services GmbH, Hammerstrasse 49, 4410 Liestal, Switzerland; (L.P.); (N.W.); (A.L.)
| | - Judith Bergs
- Department of Radiology Charité—Universitätsmedizin Berlin, CCM Charitéplatz 1, 10117 Berlin, Germany;
| | - Takashi Shimokawa
- Department of Charged Particle Therapy Research, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan; (R.H.); (N.M.); (T.I.); (S.K.); (T.S.)
- Department of Accelerator and Medical Physics, National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| |
Collapse
|
130
|
Spałek MJ, Poleszczuk J, Czarnecka AM, Dudzisz-Śledź M, Napieralska A, Matysiakiewicz J, Chojnacka M, Raciborska A, Sztuder A, Maciejczyk A, Szulc A, Skóra T, Cybulska-Stopa B, Winiecki T, Kaźmierska J, Tomasik B, Fijuth J, Rutkowski P. Radiotherapy in the Management of Pediatric and Adult Osteosarcomas: A Multi-Institutional Cohort Analysis. Cells 2021; 10:cells10020366. [PMID: 33578676 PMCID: PMC7916348 DOI: 10.3390/cells10020366] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/31/2021] [Accepted: 02/07/2021] [Indexed: 02/06/2023] Open
Abstract
Background: Due to the rarity of osteosarcoma and limited indications for radiotherapy (RT), data on RT for this tumor are scarce. This study aimed to investigate the utilization of RT for osteosarcomas in the recent 20 years and to identify factors related to patients’ response to radiation. Methods: We performed a retrospective analysis of patients irradiated for osteosarcoma treatment. We planned to assess differences in the utilization of RT between the periods of 2000–2010 and 2011–2020, identify the risk factors associated with local progression (LP), determine whether RT-related parameters are associated with LP, and calculate patients’ survival. Results: A total of 126 patients with osteosarcoma who received 181 RT treatments were identified. We found a difference in RT techniques between RT performed in the years 2000–2010 and that performed in the years 2011–2020. LP was observed after 37 (20.4%) RT treatments. Intent of RT, distant metastases, and concomitant systemic treatment affected the risk of LP. Five-year overall survival was 33% (95% confidence interval (26%–43%)). Conclusions: RT for osteosarcoma treatment has evolved from simple two-dimensional palliative irradiation into more conformal RT applied for new indications including oligometastatic and oligoprogressive disease. RT may be a valuable treatment modality for selected patients with osteosarcoma.
Collapse
Affiliation(s)
- Mateusz Jacek Spałek
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.M.C.); (M.D.-Ś.); (P.R.)
- Correspondence: ; Tel.: +48-22-546-24-55
| | - Jan Poleszczuk
- Department for Computational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-034 Warsaw, Poland;
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, 02-109 Warsaw, Poland
| | - Anna Małgorzata Czarnecka
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.M.C.); (M.D.-Ś.); (P.R.)
- Mossakowski Medical Research Centre, Department of Experimental Pharmacology, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Monika Dudzisz-Śledź
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.M.C.); (M.D.-Ś.); (P.R.)
| | - Aleksandra Napieralska
- Department of Radiotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, 44-102 Gliwice, Poland;
| | - Jacek Matysiakiewicz
- Trauma and Orthopedic Surgery Department, IXth Ward of the District Hospital of Orthopedics and Trauma Surgery in Piekary Slaskie, 41-940 Piekary Slaskie, Poland;
| | - Marzanna Chojnacka
- Department of Oncology and Radiotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, 02-034 Warsaw, Poland;
| | - Anna Raciborska
- Department of Oncology and Surgical Oncology for Children and Youth, Institute of Mother and Child, 01-211 Warsaw, Poland;
| | - Aleksandra Sztuder
- Department of Radiotherapy, Lower Silesian Oncology Centre, 53-413 Wroclaw, Poland; (A.S.); (A.M.); (A.S.)
| | - Adam Maciejczyk
- Department of Radiotherapy, Lower Silesian Oncology Centre, 53-413 Wroclaw, Poland; (A.S.); (A.M.); (A.S.)
- Department of Oncology, Faculty of Medicine, Wroclaw Medical University, 50-367 Wrocław, Poland
| | - Agata Szulc
- Department of Radiotherapy, Lower Silesian Oncology Centre, 53-413 Wroclaw, Poland; (A.S.); (A.M.); (A.S.)
| | - Tomasz Skóra
- Department of Radiotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków Branch, 31-115 Kraków, Poland;
| | - Bożena Cybulska-Stopa
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 31-115 Kraków, Poland;
| | - Tomasz Winiecki
- Radiotherapy Department II, Greater Poland Cancer Centre, 61-866 Poznan, Poland; (T.W.); (J.K.)
| | - Joanna Kaźmierska
- Radiotherapy Department II, Greater Poland Cancer Centre, 61-866 Poznan, Poland; (T.W.); (J.K.)
- Electroradiology Department, University of Medical Sciences, 61-701 Poznan, Poland
| | - Bartłomiej Tomasik
- Department of Radiotherapy, Medical University of Lodz, 92-215 Lodz, Poland; (B.T.); (J.F.)
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, 95-513 Lodz, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Jacek Fijuth
- Department of Radiotherapy, Medical University of Lodz, 92-215 Lodz, Poland; (B.T.); (J.F.)
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (A.M.C.); (M.D.-Ś.); (P.R.)
| |
Collapse
|
131
|
Cordoni F, Missiaggia M, Attili A, Welford SM, Scifoni E, La Tessa C. Generalized stochastic microdosimetric model: The main formulation. Phys Rev E 2021; 103:012412. [PMID: 33601636 PMCID: PMC7975068 DOI: 10.1103/physreve.103.012412] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/06/2021] [Indexed: 06/12/2023]
Abstract
The present work introduces a rigorous stochastic model, called the generalized stochastic microdosimetric model (GSM^{2}), to describe biological damage induced by ionizing radiation. Starting from the microdosimetric spectra of energy deposition in tissue, we derive a master equation describing the time evolution of the probability density function of lethal and potentially lethal DNA damage induced by a given radiation to a cell nucleus. The resulting probability distribution is not required to satisfy any a priori conditions. After the initial assumption of instantaneous irradiation, we generalized the master equation to consider damage induced by a continuous dose delivery. In addition, spatial features and damage movement inside the nucleus have been taken into account. In doing so, we provide a general mathematical setting to fully describe the spatiotemporal damage formation and evolution in a cell nucleus. Finally, we provide numerical solutions of the master equation exploiting Monte Carlo simulations to validate the accuracy of GSM^{2}. Development of GSM^{2} can lead to improved modeling of radiation damage to both tumor and normal tissues, and thereby impact treatment regimens for better tumor control and reduced normal tissue toxicities.
Collapse
Affiliation(s)
- F Cordoni
- Department of Computer Science, University of Verona, Verona, Italy and TIFPA-INFN, Trento, Italy
| | - M Missiaggia
- Department of Physics, University of Trento, Trento, Italy and TIFPA-INFN, Trento, Italy
| | | | - S M Welford
- Department of Radiation Oncology, University of Miami, Miller School of Medicine, Miami, Florida 33136, USA
| | | | - C La Tessa
- Department of Physics, University of Trento, Trento, Italy and TIFPA - INFN, Trento, Italy
| |
Collapse
|
132
|
Beltran C, Amos RA, Rong Y. We are ready for clinical implementation of Carbon Ion Radiotherapy in the United States. J Appl Clin Med Phys 2020; 21:6-9. [PMID: 33319499 PMCID: PMC7769388 DOI: 10.1002/acm2.13133] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 12/24/2022] Open
Affiliation(s)
- Chris Beltran
- Department of Radiation Oncology, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | - Richard A Amos
- Proton and Advanced Radiotherapy Group, Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Yi Rong
- Department of Radiation Oncology, Mayo Clinic Arizona, Phoenix, AZ, USA
| |
Collapse
|
133
|
Kumari S, Mukherjee S, Sinha D, Abdisalaam S, Krishnan S, Asaithamby A. Immunomodulatory Effects of Radiotherapy. Int J Mol Sci 2020; 21:E8151. [PMID: 33142765 PMCID: PMC7663574 DOI: 10.3390/ijms21218151] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023] Open
Abstract
Radiation therapy (RT), an integral component of curative treatment for many malignancies, can be administered via an increasing array of techniques. In this review, we summarize the properties and application of different types of RT, specifically, conventional therapy with x-rays, stereotactic body RT, and proton and carbon particle therapies. We highlight how low-linear energy transfer (LET) radiation induces simple DNA lesions that are efficiently repaired by cells, whereas high-LET radiation causes complex DNA lesions that are difficult to repair and that ultimately enhance cancer cell killing. Additionally, we discuss the immunogenicity of radiation-induced tumor death, elucidate the molecular mechanisms by which radiation mounts innate and adaptive immune responses and explore strategies by which we can increase the efficacy of these mechanisms. Understanding the mechanisms by which RT modulates immune signaling and the key players involved in modulating the RT-mediated immune response will help to improve therapeutic efficacy and to identify novel immunomodulatory drugs that will benefit cancer patients undergoing targeted RT.
Collapse
Affiliation(s)
- Sharda Kumari
- Division of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (S.K.); (D.S.); (S.A.)
| | - Shibani Mukherjee
- Division of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (S.K.); (D.S.); (S.A.)
| | - Debapriya Sinha
- Division of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (S.K.); (D.S.); (S.A.)
| | - Salim Abdisalaam
- Division of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (S.K.); (D.S.); (S.A.)
| | - Sunil Krishnan
- Department of Radiation Oncology, Mayo Clinic Florida, Jacksonville, FL 32224, USA;
| | - Aroumougame Asaithamby
- Division of Molecular Radiation Biology, Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (S.K.); (D.S.); (S.A.)
| |
Collapse
|