1
|
Billoir M, Crepin D, Plaszczynski S, Grammaticos B, Seksek O, Badoual M. The temporal response of a glioma cell population to irradiation: modelling the effect of dose and cell density. ROYAL SOCIETY OPEN SCIENCE 2025; 12:241917. [PMID: 40309193 PMCID: PMC12040453 DOI: 10.1098/rsos.241917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/09/2025] [Accepted: 02/10/2025] [Indexed: 05/02/2025]
Abstract
Time-lapse fluorescence microscopy experiments were performed to track the cell density of F98 glioma cells under varying radiation doses and initial cell densities. Based on these results, a compartmental model characterizing the temporal response of a cancerous cell population to single-dose radiation therapy was developed. This model reproduces very well all the experimental data, with only three free parameters (and four others that are fixed). It allows us to have access and follow the evolution of different cell populations after irradiation, in particular, the senescent and repaired cell populations. From these different cell populations, surviving fractions could also be estimated. Most importantly, our model allows us to analyze and quantify an inhibition effect (or cohort effect) of the dead and senescent cell populations on the regrowth of the repaired one.
Collapse
Affiliation(s)
- Marianne Billoir
- CNRS/IN2P3, IJCLab, Université Paris-Saclay, Orsay, France
- IJCLab, Université Paris Cité, OrsayÎle-de-France, France
| | - Delphine Crepin
- CNRS/IN2P3, IJCLab, Université Paris-Saclay, Orsay, France
- IJCLab, Université Paris Cité, OrsayÎle-de-France, France
| | - Stéphane Plaszczynski
- CNRS/IN2P3, IJCLab, Université Paris-Saclay, Orsay, France
- IJCLab, Université Paris Cité, OrsayÎle-de-France, France
| | - Basile Grammaticos
- CNRS/IN2P3, IJCLab, Université Paris-Saclay, Orsay, France
- IJCLab, Université Paris Cité, OrsayÎle-de-France, France
| | - Olivier Seksek
- CNRS/IN2P3, IJCLab, Université Paris-Saclay, Orsay, France
- IJCLab, Université Paris Cité, OrsayÎle-de-France, France
| | - Mathilde Badoual
- CNRS/IN2P3, IJCLab, Université Paris-Saclay, Orsay, France
- IJCLab, Université Paris Cité, OrsayÎle-de-France, France
| |
Collapse
|
2
|
Terashima S, Tatemura R, Saito W, Hosokawa Y. Evaluation of the influence of radiation-induced cohort effect in cell populations receiving different doses. Int J Radiat Biol 2025; 101:341-350. [PMID: 39899278 DOI: 10.1080/09553002.2025.2459086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 02/04/2025]
Abstract
PURPOSE A non-targeted effect called radiation-induced cohort effect, which results in interactions among irradiated neighboring cells through cellular communication, has been reported. In high-precision radiotherapy, the dose is localized to the tumor, and rapid spatial changes occur in dose distribution. However, the effect of irradiating a population of cells with non-uniform doses remains unknown. In this study, we evaluated the influence of cohort effect by creating cell populations irradiated with different doses using human oral squamous cell carcinoma (SAS) and human lung (A549) cells. MATERIALS AND METHODS Cell populations irradiated with different doses were created in two ways: direct contact co-culture (DCC) using a cell tracer dye and indirect contact co-culture (ICC) using cell culture inserts to assess the effects of soluble factors. Target cells were irradiated with 4 Gy and co-cultured cells with 0, 0.8, 3.2, and 4 Gy. In DCC, cell proliferation assays were performed using a flow cytometer, and in ICC, modified high-density survival, clonogenic, and apoptosis assays were performed. RESULTS In DCC, irradiation of co-cultured cells with X-rays increased the relative proliferation rate of the target cells. Similarly, irradiating co-cultured cells using ICC with X-rays increased the relative survival rate of target cells. CONCLUSIONS The results of this study showed that, even if there is a sharp decrease in dose near the tumor, the cytocidal effect on the tumor is not adversely affected. In addition, soluble factors were found to be involved in cohort effect.
Collapse
Affiliation(s)
- Shingo Terashima
- Department of Radiation Science, Hirosaki University Graduate School of Health Sciences, Hirosaki, Japan
| | - Ryota Tatemura
- Department of Radiology, Division of Medical Technology, Hirosaki University School of Medicine and Hospital, Hirosaki, Japan
| | - Wataru Saito
- Plant Operation Department, Reprocessing Plant, Reprocessing Business Division, Japan Nuclear Fuel Limited, Rokkasho-mura, Japan
| | - Yoichiro Hosokawa
- Department of Rehabilitation Sciences, Hirosaki University of Health and Welfare, Hirosaki, Japan
| |
Collapse
|
3
|
Katugampola S, Wang J, Howell RW. MIRD Pamphlet No. 31: MIRDcell V4-Artificial Intelligence Tools to Formulate Optimized Radiopharmaceutical Cocktails for Therapy. J Nucl Med 2024; 65:1965-1973. [PMID: 39448267 PMCID: PMC11619582 DOI: 10.2967/jnumed.123.267238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 09/18/2024] [Indexed: 10/26/2024] Open
Abstract
Radiopharmaceutical cocktails have been developed over the years to treat cancer. Cocktails of agents are attractive because 1 radiopharmaceutical is unlikely to have the desired therapeutic effect because of nonuniform uptake by the targeted cells. Therefore, multiple radiopharmaceuticals targeting different receptors on a cell is warranted. However, past implementations in vivo have not met with convincing results because of the absence of optimization strategies. Here we present artificial intelligence (AI) tools housed in a new version of our software platform, MIRDcell V4, that optimize a cocktail of radiopharmaceuticals by minimizing the total disintegrations needed to achieve a given surviving fraction (SF) of tumor cells. Methods: AI tools are developed within MIRDcell V4 using an optimizer based on the sequential least-squares programming algorithm. The algorithm determines the molar activities for each drug in the cocktail that minimize the total disintegrations required to achieve a specified SF. Tools are provided for populations of cells that do not cross-irradiate (e.g., circulating or disseminated tumor cells) and for multicellular clusters (e.g., micrometastases). The tools were tested using model data, flow cytometry data for suspensions of single cells labeled with fluorochrome-labeled antibodies, and 3-dimensional spatiotemporal kinetics in spheroids for fluorochrome-loaded liposomes. Results: Experimental binding distributions of 4 211At-antibodies were considered for treating suspensions of MDA-MB-231 human breast cancer cells. A 2-drug combination reduced the number of 211At decays required by a factor of 1.6 relative to the best single antibody. In another study, 2 radiopharmaceuticals radiolabeled with 195mPt were each distributed lognormally in a hypothetical multicellular cluster. Here, the 2-drug combination required 1.7-fold fewer decays than did either drug alone. Finally, 2 225Ac-labeled drugs that provide different radial distributions within a spheroid require about one half of the disintegrations required by the best single agent. Conclusion: The MIRDcell AI tools determine optimized drug combinations and corresponding molar activities needed to achieve a given SF. This approach could be used to analyze a sample of cells obtained from cell culture, animal, or patient to predict the best combination of drugs for maximum therapeutic effect with the least total disintegrations.
Collapse
Affiliation(s)
- Sumudu Katugampola
- Division of Radiation Research, Department of Radiology, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Jianchao Wang
- Division of Radiation Research, Department of Radiology, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Roger W Howell
- Division of Radiation Research, Department of Radiology, New Jersey Medical School, Rutgers University, Newark, New Jersey
| |
Collapse
|
4
|
Moscardini-Martelli J, Rodríguez-Camacho A, Torres-Ríos JA, Meraz-Soto JM, Flores-Vázquez JG, Hernández-Sánchez LC, Lozano-Ruiz FJ, Maldonado-Magos F, Cid-Sánchez D, Flores-Balcázar CH, Celis-López MÁ, Gutiérrez-Aceves GA, Flores-Vázquez F, Moreno-Jiménez S. A Comprehensive Revision of Radiation Immunotherapy and the Abscopal Effect in Central Nervous System Metastases: Reassessing the Frontier. Curr Issues Mol Biol 2024; 46:11075-11085. [PMID: 39451538 PMCID: PMC11506806 DOI: 10.3390/cimb46100658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 10/26/2024] Open
Abstract
Seventy years ago, Robin Mole introduced the concept of the abscopal effect to describe a rare phenomenon. This occurs when local radiation triggers an immune-mediated reduction in tumors outside the treated area but within the same organism. Observing this effect has been linked to improved overall and progression-free survival in patients who experience it. While the abscopal effect was once considered rare, it is now being observed more frequently due to the combination of radiation with immunotherapy. As a result, more researchers are exploring this study area, which shows promise for excellent results. This review focuses explicitly on the immunological implications of activating the abscopal effect through ionizing radiation in the central nervous system and explores the potentially involved immunological pathways.
Collapse
Affiliation(s)
- Júlia Moscardini-Martelli
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - Alejandro Rodríguez-Camacho
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
- Radiotherapy Service, National Cancer Institute, Mexico City 14080, Mexico; (F.J.L.-R.); (F.M.-M.); (D.C.-S.); (C.H.F.-B.)
| | - Jorge Alejandro Torres-Ríos
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - Juan Marcos Meraz-Soto
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - José Guillermo Flores-Vázquez
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - Laura Crystell Hernández-Sánchez
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - Francisco Javier Lozano-Ruiz
- Radiotherapy Service, National Cancer Institute, Mexico City 14080, Mexico; (F.J.L.-R.); (F.M.-M.); (D.C.-S.); (C.H.F.-B.)
| | - Federico Maldonado-Magos
- Radiotherapy Service, National Cancer Institute, Mexico City 14080, Mexico; (F.J.L.-R.); (F.M.-M.); (D.C.-S.); (C.H.F.-B.)
| | - Dharely Cid-Sánchez
- Radiotherapy Service, National Cancer Institute, Mexico City 14080, Mexico; (F.J.L.-R.); (F.M.-M.); (D.C.-S.); (C.H.F.-B.)
| | | | - Miguel Ángel Celis-López
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - Guillermo Axayacatl Gutiérrez-Aceves
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | | | - Sergio Moreno-Jiménez
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
- American British Cowdray Medical Center, Neurological Center, Mexico City 01120, Mexico
| |
Collapse
|
5
|
Mansur A, Habibollahi P, Fang A, Mahvash A, Etezadi V, Liddell RP, Camacho JC, Cohen EI, Kokabi N, Arepally A, Georgiades C, Nezami N. New frontiers in radioembolization. Ther Adv Med Oncol 2024; 16:17588359241280692. [PMID: 39371617 PMCID: PMC11456171 DOI: 10.1177/17588359241280692] [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: 01/17/2024] [Accepted: 08/19/2024] [Indexed: 10/08/2024] Open
Abstract
Radioembolization is a locoregional transarterial therapy that combines radionuclide and micron-sized beads to deliver radiation internally to the target tumors based on the arterial blood flow. While initially developed as a palliative treatment option, radioembolization is now used for curative intent treatment, neoadjuvant therapy, and method to downstage or bridge for liver transplant. Radioembolization has become increasingly utilized and is an important therapeutic option for the management of hepatocellular carcinoma and liver metastasis. This article provides an overview of the techniques, challenges, and novel developments in radioembolization, including new dosimetry techniques, radionuclides, and new target tumors.
Collapse
Affiliation(s)
| | - Peiman Habibollahi
- Division of Diagnostic Imaging, Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Adam Fang
- Division of Vascular and Interventional Radiology, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Armeen Mahvash
- Division of Diagnostic Imaging, Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vahid Etezadi
- Division of Vascular and Interventional Radiology, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Robert P. Liddell
- Division of Vascular and Interventional Radiology, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Juan C. Camacho
- Department of Clinical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
- Vascular and Interventional Radiology, Radiology Associates of Florida, Sarasota, FL, USA
| | - Emil I. Cohen
- Division of Vascular and Interventional Radiology, Department of Radiology, Georgetown University School of Medicine, Washington, DC, USA
| | - Nima Kokabi
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Aravind Arepally
- Radiology Associates of Atlanta, Atlanta, GA, USA
- ABK Biomedical Inc., Atlanta, GA, USA
| | - Christos Georgiades
- Division of Vascular and Interventional Radiology, Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nariman Nezami
- Division of Vascular and Interventional Radiology, Department of Radiology, Georgetown University School of Medicine, 3800 Reservoir Road, NW, CCC Bldg., Room CG225, Washington, DC 20007, USA
| |
Collapse
|
6
|
Gardner LL, Thompson SJ, O'Connor JD, McMahon SJ. Modelling radiobiology. Phys Med Biol 2024; 69:18TR01. [PMID: 39159658 DOI: 10.1088/1361-6560/ad70f0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/19/2024] [Indexed: 08/21/2024]
Abstract
Radiotherapy has played an essential role in cancer treatment for over a century, and remains one of the best-studied methods of cancer treatment. Because of its close links with the physical sciences, it has been the subject of extensive quantitative mathematical modelling, but a complete understanding of the mechanisms of radiotherapy has remained elusive. In part this is because of the complexity and range of scales involved in radiotherapy-from physical radiation interactions occurring over nanometres to evolution of patient responses over months and years. This review presents the current status and ongoing research in modelling radiotherapy responses across these scales, including basic physical mechanisms of DNA damage, the immediate biological responses this triggers, and genetic- and patient-level determinants of response. Finally, some of the major challenges in this field and potential avenues for future improvements are also discussed.
Collapse
Affiliation(s)
- Lydia L Gardner
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7AE, United Kingdom
| | - Shannon J Thompson
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7AE, United Kingdom
| | - John D O'Connor
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7AE, United Kingdom
- Ulster University School of Engineering, York Street, Belfast BT15 1AP, United Kingdom
| | - Stephen J McMahon
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7AE, United Kingdom
| |
Collapse
|
7
|
Martin OA, Sykes PJ, Lavin M, Engels E, Martin RF. What's Changed in 75 Years of RadRes? - An Australian Perspective on Selected Topics. Radiat Res 2024; 202:309-327. [PMID: 38966925 DOI: 10.1667/rade-24-00037.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/09/2024] [Indexed: 07/06/2024]
Abstract
Several scientific themes are reviewed in the context of the 75-year period relevant to this special platinum issue of Radiation Research. Two criteria have been considered in selecting the scientific themes. One is the exposure of the associated research activity in the annual meetings of the Radiation Research Society (RRS) and in the publications of the Society's Journal, thus reflecting the interest of members of RRS. The second criteria is a focus on contributions from Australian members of RRS. The first theme is the contribution of radiobiology to radiation oncology, featuring two prominent Australian radiation oncologists, the late Rod Withers and his younger colleague, Lester Peters. Two other themes are also linked to radiation oncology; preclinical research aimed at developing experimental radiotherapy modalities, namely microbeam radiotherapy (MRT) and Auger endoradiotherapy. The latter has a long history, in contrast to MRT, especially in Australia, given that the associated medical beamline at the Australian Synchrotron in Melbourne only opened in 2011. Another theme is DNA repair, which has a trajectory parallel to the 75-year period of interest, given the birth of molecular biology in the 1950s. The low-dose radiobiology theme has a similar timeline, predominantly prompted by the nuclear era, which is also connected to the radioprotector theme, although radioprotectors also have a long-established potential utility in cancer radiotherapy. Finally, two themes are associated with biodosimetry. One is the micronucleus assay, highlighting the pioneering contribution from Michael Fenech in Adelaide, South Australia, and the other is the γ-H2AX assay and its widespread clinical applications.
Collapse
Affiliation(s)
- Olga A Martin
- Centre of Medical Radiation Physics (CMRP), University of Wollongong, Wollongong, NSW, Australia
| | - Pamela J Sykes
- College of Medicine and Public Health, Flinders University and Medical Centre, Bedford Park, SA, Australia
| | - Martin Lavin
- Centre for Clinical Research, University of Queensland, QSL, Brisbane, Australia
| | - Elette Engels
- Centre of Medical Radiation Physics (CMRP), University of Wollongong, Wollongong, NSW, Australia
- Australian Synchrotron, Australian Nuclear Science and Technology Organisation (ANSTO), Clayton, VIC, Australia
| | - Roger F Martin
- School of Chemistry, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
8
|
Jadhav TS, Sansare K, Sreenivasan V, Unnikrishnan A, Vahanwala S. A systematic review and meta-analysis of the genotoxic and cytotoxic effects on oral epithelium induced by cone beam computed tomography. Oral Surg Oral Med Oral Pathol Oral Radiol 2024; 138:324-334. [PMID: 38570275 DOI: 10.1016/j.oooo.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 02/13/2024] [Accepted: 03/03/2024] [Indexed: 04/05/2024]
Abstract
OBJECTIVE To evaluate the occurrence of genotoxic and cytotoxic effects in oral epithelium after exposure of patients to cone beam computed tomography (CBCT). METHODS A systematic review (SR) was conducted following the PECO (Population, Exposure, Comparison, Outcome) criteria. The study was registered in the International Prospective Register of Systematic Reviews (PROSPERO). A search was performed on the PubMed, Scopus, ScienceDirect, and Google Scholar databases. Effect size and heterogeneity of data were evaluated statistically. The Joanna Briggs Institute questionnaire for observational studies was utilized to assess the risk of bias. The GRADE tool was applied for the assessment of the quality of evidence. Begg's funnel plot was used to evaluate publication bias. RESULTS In total, 10 full-text articles were included in the SR, with 6 of them in the meta-analysis. The SR showed a significant increase in micronuclei after exposure, with a large effect size of 1.03. For genotoxicity, the tau2 for heterogeneity was 0.96, the chi-squared test for heterogeneity P < .00001, the I2 statistics for random effects was 91%, and the overall effect for Z value was 2.46 (P = .01). The risk of bias was low, the quality of evidence was strong, and publication bias was absent. CONCLUSION CBCT can cause genotoxicity in the oral epithelium with a large effect size. The measure of cytotoxicity after CBCT exposure was not possible due to the lack of homogeneity of the included articles.
Collapse
Affiliation(s)
- Tanushree S Jadhav
- Oral Medicine and Maxillofacial Radiology, Nair Hospital Dental College, Mumbai, India.
| | - Kaustubh Sansare
- Oral Medicine and Maxillofacial Radiology, Nair Hospital Dental College, Mumbai, India
| | - Venkatraman Sreenivasan
- Oral Medicine and Maxillofacial Radiology, Bharathi Vidyapeeth Dental College and Hospital, Navi Mumbai, India
| | - Aswathi Unnikrishnan
- Oral Medicine and Maxillofacial Radiology, Nair Hospital Dental College, Mumbai, India
| | - Sonal Vahanwala
- Oral Medicine and Maxillofacial Radiology, Nair Hospital Dental College, Mumbai, India
| |
Collapse
|
9
|
Lu Q, Yan W, Zhu A, Tubin S, Mourad WF, Yang J. Combining spatially fractionated radiation therapy (SFRT) and immunotherapy opens new rays of hope for enhancing therapeutic ratio. Clin Transl Radiat Oncol 2024; 44:100691. [PMID: 38033759 PMCID: PMC10684810 DOI: 10.1016/j.ctro.2023.100691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/04/2023] [Accepted: 10/15/2023] [Indexed: 12/02/2023] Open
Abstract
Spatially Fractionated Radiation Therapy (SFRT) is a form of radiotherapy that delivers a single large dose of radiation within the target volume in a heterogeneous pattern with regions of peak dosage and regions of under dosage. SFRT types can be defined by how the heterogeneous pattern of radiation is obtained. Immune checkpoint inhibitors (ICIs) have been approved for various malignant tumors and are widely used to treat patients with metastatic cancer. The efficacy of ICI monotherapy is limited due to the "cold" tumor microenvironment. Fractionated radiotherapy can achieve higher doses per fraction to the target tumor, and induce immune activation (immodulate tumor immunogenicity and microenvironment). Therefore, coupling ICI therapy and fractionated radiation therapy could significantly improve the outcome of metastatic cancer. This review focuses on both preclinical and clinical studies that use a combination of radiotherapy and ICI therapy in cancer.
Collapse
Affiliation(s)
- Qiuxia Lu
- Foshan Fosun Chancheng Hospital, P.R. China
- Junxin Precision Oncology Group, P.R. China
| | - Weisi Yan
- Baptist Health System, Lexington, KY, United States
- Junxin Precision Oncology Group, P.R. China
| | - Alan Zhu
- Mayo Clinic Alix School of Medicine, Scottsdale, AZ, United States
| | - Slavisa Tubin
- Albert Einstein Collage of Medicine New York, Center for Ion Therapy, Medaustron, Austria
| | - Waleed F. Mourad
- Department of Radiation Medicine Markey Cancer Center, University of Kentucky - College of Medicine, United States
| | - Jun Yang
- Foshan Fosun Chancheng Hospital, P.R. China
- Junxin Precision Oncology Group, P.R. China
| |
Collapse
|
10
|
Khazaei Monfared Y, Heidari P, Klempner SJ, Mahmood U, Parikh AR, Hong TS, Strickland MR, Esfahani SA. DNA Damage by Radiopharmaceuticals and Mechanisms of Cellular Repair. Pharmaceutics 2023; 15:2761. [PMID: 38140100 PMCID: PMC10748326 DOI: 10.3390/pharmaceutics15122761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
DNA is an organic molecule that is highly vulnerable to chemical alterations and breaks caused by both internal and external factors. Cells possess complex and advanced mechanisms, including DNA repair, damage tolerance, cell cycle checkpoints, and cell death pathways, which together minimize the potentially harmful effects of DNA damage. However, in cancer cells, the normal DNA damage tolerance and response processes are disrupted or deregulated. This results in increased mutagenesis and genomic instability within the cancer cells, a known driver of cancer progression and therapeutic resistance. On the other hand, the inherent instability of the genome in rapidly dividing cancer cells can be exploited as a tool to kill by imposing DNA damage with radiopharmaceuticals. As the field of targeted radiopharmaceutical therapy (RPT) is rapidly growing in oncology, it is crucial to have a deep understanding of the impact of systemic radiation delivery by radiopharmaceuticals on the DNA of tumors and healthy tissues. The distribution and activation of DNA damage and repair pathways caused by RPT can be different based on the characteristics of the radioisotope and molecular target. Here we provide a comprehensive discussion of the biological effects of RPTs, with the main focus on the role of varying radioisotopes in inducing direct and indirect DNA damage and activating DNA repair pathways.
Collapse
Affiliation(s)
- Yousef Khazaei Monfared
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (Y.K.M.); (P.H.); (U.M.)
| | - Pedram Heidari
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (Y.K.M.); (P.H.); (U.M.)
| | - Samuel J. Klempner
- Division of Hematology-Oncology, Department of Medicine, Mass General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (S.J.K.); (A.R.P.); (M.R.S.)
| | - Umar Mahmood
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (Y.K.M.); (P.H.); (U.M.)
| | - Aparna R. Parikh
- Division of Hematology-Oncology, Department of Medicine, Mass General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (S.J.K.); (A.R.P.); (M.R.S.)
| | - Theodore S. Hong
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA;
| | - Matthew R. Strickland
- Division of Hematology-Oncology, Department of Medicine, Mass General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (S.J.K.); (A.R.P.); (M.R.S.)
| | - Shadi A. Esfahani
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (Y.K.M.); (P.H.); (U.M.)
| |
Collapse
|
11
|
Liepe K. Pros and Cons of Alpha versus Beta Bone Seeking Agents in the Treatment of Cancer Pain. World J Nucl Med 2023; 22:255-256. [PMID: 38152106 PMCID: PMC10751140 DOI: 10.1055/s-0043-1774731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023] Open
Affiliation(s)
- Knut Liepe
- Department of Nuclear Medicine, Klinikum Frankfurt (Oder), Brandenburg, Germany
| |
Collapse
|
12
|
Maier A, Bailey T, Hinrichs A, Lerchl S, Newman RT, Fournier C, Vandevoorde C. Experimental Setups for In Vitro Studies on Radon Exposure in Mammalian Cells-A Critical Overview. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:ijerph20095670. [PMID: 37174189 PMCID: PMC10178159 DOI: 10.3390/ijerph20095670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023]
Abstract
Naturally occurring radon and its short lived progeny are the second leading cause of lung cancer after smoking, and the main risk factor for non-smokers. The radon progeny, mainly Polonium-218 (218Po) and Polonium-214 (214Po), are responsible for the highest dose deposition in the bronchial epithelium via alpha-decay. These alpha-particles release a large amount of energy over a short penetration range, which results in severe and complex DNA damage. In order to unravel the underlying biological mechanisms which are triggered by this complex DNA damage and eventually give rise to carcinogenesis, in vitro radiobiology experiments on mammalian cells have been performed using radon exposure setups, or radon analogues, which mimic alpha-particle exposure. This review provides an overview of the different experimental setups, which have been developed and used over the past decades for in vitro radon experiments. In order to guarantee reliable results, the design and dosimetry of these setups require careful consideration, which will be emphasized in this work. Results of these in vitro experiments, particularly on bronchial epithelial cells, can provide valuable information on biomarkers, which can assist to identify exposures, as well as to study the effects of localized high dose depositions and the heterogeneous dose distribution of radon.
Collapse
Affiliation(s)
- Andreas Maier
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, 64291 Darmstadt, Germany
| | - Tarryn Bailey
- Department of Physics, Stellenbosch University, Stellenbosch, Cape Town 7600, South Africa
- Radiation Biophysics Division, Separated Sector Cyclotron Laboratory, NRF-iThemba LABS, Cape Town 7129, South Africa
| | - Annika Hinrichs
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, 64291 Darmstadt, Germany
- Physics Department, Goethe University Frankfurt am Main, 60438 Frankfurt am Main, Germany
| | - Sylvie Lerchl
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, 64291 Darmstadt, Germany
| | - Richard T Newman
- Department of Physics, Stellenbosch University, Stellenbosch, Cape Town 7600, South Africa
| | - Claudia Fournier
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, 64291 Darmstadt, Germany
| | - Charlot Vandevoorde
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, 64291 Darmstadt, Germany
- Radiation Biophysics Division, Separated Sector Cyclotron Laboratory, NRF-iThemba LABS, Cape Town 7129, South Africa
| |
Collapse
|
13
|
Pakniyat F, Mozdarani H, Nedaie HA, Mahmoudzadeh A, Salimi M, Gholami S. Bystander Response Following High-Dose X-irradiation; Time-dependent Nature of GammaH2AX Foci and Cell Death Consequences. J Biomed Phys Eng 2023; 13:17-28. [PMID: 36818004 PMCID: PMC9923241 DOI: 10.31661/jbpe.v0i0.2001-1053] [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: 01/19/2020] [Accepted: 05/07/2020] [Indexed: 01/31/2023]
Abstract
Background The paradigm shifts in target theory could be defined as the radiation-triggered bystander response in which the radiation deleterious effects occurred in the adjacent cells. Objective This study aims to assess bystander response in terms of DNA damage and their possible cell death consequences following high-dose radiotherapy. Temporal characteristics of gH2AX foci as a manifestation of DNA damage were also evaluated. Material and Methods In this experimental study, bystander response was investigated in human carcinoma cells of HeLa and HN5, neighboring those that received high doses. Medium transfer was performed from 10 Gy-irradiated donors to 1.5 Gy-irradiated recipients. GammaH2AX foci, clonogenic and apoptosis assays were investigated. The gH2AX foci time-point study was implemented 1, 4, and 24 h after the medium exchange. Results DNA damage was enhanced in HeLa and HN5 bystander cells with the ratio of 1.27 and 1.72, respectively, which terminated in more than two-fold clonogenic survival decrease, along with gradual apoptosis increase. GammH2AX foci temporal characterization revealed maximum foci scoring at the 1 h time-point in HeLa, and also 4 h in HN5, which remained even 24 h after the medium sharing in higher level than the control group. Conclusion The time-dependent nature of bystander-induced gH2AX foci as a DNA damage surrogate marker was highlighted with the persistent foci at 24 h. considering an outcome of bystander-induced DNA damage, predominant role of clonogenic cell death was also elicited compared to apoptosis. Moreover, the role of high-dose bystander response observed in the current work clarified bystander potential implications in radiotherapy.
Collapse
Affiliation(s)
- Fatemeh Pakniyat
- Department of Medical Physics and Biomedical Engineering, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Mozdarani
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hassan Ali Nedaie
- Department of Medical Physics and Biomedical Engineering, Tehran University of Medical Sciences, Tehran, Iran
- Radiation Oncology Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Aziz Mahmoudzadeh
- Department of Bioscience and Biotechnology, Malek-Ashtar University of Technology, Tehran, Iran
| | - Mahdieh Salimi
- Department of Medical Genetics, Medical Biotechnology Institute, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Somayeh Gholami
- Radiation Oncology Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Hara D, Tao W, Schmidt RM, Yang YP, Daunert S, Dogan N, Ford JC, Pollack A, Shi J. Boosted Radiation Bystander Effect of PSMA-Targeted Gold Nanoparticles in Prostate Cancer Radiosensitization. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:4440. [PMID: 36558293 PMCID: PMC9784958 DOI: 10.3390/nano12244440] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 06/17/2023]
Abstract
Metal nanoparticles are effective radiosensitizers that locally enhance radiation doses in targeted cancer cells. Compared with other metal nanoparticles, gold nanoparticles (GNPs) exhibit high biocompatibility, low toxicity, and they increase secondary electron scatter. Herein, we investigated the effects of active-targeting GNPs on the radiation-induced bystander effect (RIBE) in prostate cancer cells. The impact of GNPs on the RIBE presents implications for secondary cancers or spatially fractionated radiotherapy treatments. Anti-prostate-specific membrane antigen (PSMA) antibodies were conjugated with PEGylated GNPs through EDC-NHS chemistry. The media transfer technique was performed to induce the RIBE on the non-irradiated bystander cells. This study focused on the LNCaP cell line, because it can model a wide range of stages relating to prostate cancer progression, including the transition from androgen dependence to castration resistance and bone metastasis. First, LNCaP cells were pretreated with phosphate buffered saline (PBS) or PSMA-targeted GNPs (PGNPs) for 24 h and irradiated with 160 kVp X-rays (0-8 Gy). Following that, the collected culture media were filtered (sterile 0.45 µm polyethersulfone) in order to acquire PBS- and PGNP- conditioned media (CM). Then, PBS- and PGNP-CM were transferred to the bystander cells that were loaded with/without PGNPs. MTT, γ-H2AX, clonogenic assays and reactive oxygen species assessments were performed to compare RIBE responses under different treatments. Compared with 2 Gy-PBS-CM, 8 Gy-PBS-CM demonstrated a much higher RIBE response, thus validating the dose dependence of RIBE in LNCaP cells. Compared with PBS-CM, PGNP-CM exhibited lower cell viability, higher DNA damage, and a smaller survival fraction. In the presence of PBS-CM, bystander cells loaded with PGNPs showed increased cell death compared with cells that did not have PGNPs. These results demonstrate the PGNP-boosted expression and sensitivity of RIBE in prostate cancer cells.
Collapse
Affiliation(s)
- Daiki Hara
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Biomedical Engineering, College of Engineering, University of Miami, Miami, FL 33146, USA
| | - Wensi Tao
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Ryder M. Schmidt
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Biomedical Engineering, College of Engineering, University of Miami, Miami, FL 33146, USA
| | - Yu-Ping Yang
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Sylvia Daunert
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Nesrin Dogan
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Biomedical Engineering, College of Engineering, University of Miami, Miami, FL 33146, USA
| | - John Chetley Ford
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Biomedical Engineering, College of Engineering, University of Miami, Miami, FL 33146, USA
| | - Alan Pollack
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Junwei Shi
- Department of Radiation Oncology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
15
|
Uchida H, Ingalls MH, Maruyama EO, Johnston CJ, Hernady E, Faustoferri RC, Ovitt CE. Short-term and bystander effects of radiation on murine submandibular glands. Dis Model Mech 2022; 15:dmm049570. [PMID: 36263624 PMCID: PMC9683099 DOI: 10.1242/dmm.049570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 10/13/2022] [Indexed: 11/20/2022] Open
Abstract
Many patients treated for head and neck cancers experience salivary gland hypofunction due to radiation damage. Understanding the mechanisms of cellular damage induced by radiation treatment is important in order to design methods of radioprotection. In addition, it is crucial to recognize the indirect effects of irradiation and the systemic responses that may alter saliva secretion. In this study, radiation was delivered to murine submandibular glands (SMGs) bilaterally, using a 137Cs gamma ray irradiator, or unilaterally, using a small-animal radiation research platform (SARRP). Analysis at 3, 24 and 48 h showed dynamic changes in mRNA and protein expression in SMGs irradiated bilaterally. Unilateral irradiation using the SARRP caused similar changes in the irradiated SMGs, as well as significant off-target, bystander effects in the non-irradiated contralateral SMGs.
Collapse
Affiliation(s)
- Hitoshi Uchida
- Center for Oral Biology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Matthew H. Ingalls
- Center for Oral Biology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Eri O. Maruyama
- Center for Oral Biology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Carl J. Johnston
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Eric Hernady
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Roberta C. Faustoferri
- Center for Oral Biology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Catherine E. Ovitt
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642USA
| |
Collapse
|
16
|
Changes in Radiosensitivity to Gamma-Rays of Lymphocytes from Hyperthyroid Patients Treated with I-131. Int J Mol Sci 2022; 23:ijms231710156. [PMID: 36077557 PMCID: PMC9456272 DOI: 10.3390/ijms231710156] [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: 07/18/2022] [Revised: 08/17/2022] [Accepted: 08/27/2022] [Indexed: 11/17/2022] Open
Abstract
This study investigated the peripheral blood lymphocytes (PBL) response to a dose of γ-rays in patients treated with radioiodine (I-131) for hyperthyroidism vs. healthy controls, to gain information about the individual lymphocytes’ radio-sensitivity. Blood samples were taken from 18 patients and 10 healthy donors. Phosphorylated histone variant H2AX (γ-H2AX) and micronuclei (MN) induction were used to determine the change in PBL radio-sensitivity and the correlations between the two types of damage. The two assays showed large inter-individual variability in PBL background damage and in radio-sensitivity (patients vs. healthy donors). In particular, they showed an increased radio-sensitivity in 36% and 33% of patients, decrease in 36% and 44%, respectively. There was a scarce correlation between the two assays and no dependence on age or gender. A significant association was found between high radio-sensitivity conditions and induced hypothyroidism. PBL radio-sensitivity in the patient group was not significantly affected by treatment with I-131, whereas there were significant changes inter-individually. The association found between clinical response and PBL radio-sensitivity suggests that the latter could be used in view of the development of personalized treatments.
Collapse
|
17
|
Katugampola S, Wang J, Rosen A, Howell RW. MIRD Pamphlet No. 27: MIRDcell V3, a Revised Software Tool for Multicellular Dosimetry and Bioeffect Modeling. J Nucl Med 2022; 63:1441-1449. [PMID: 35145016 PMCID: PMC9454469 DOI: 10.2967/jnumed.121.263253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/26/2022] [Indexed: 01/26/2023] Open
Abstract
Radiopharmaceutical therapy is growing rapidly. However, yet to be addressed is the implementation of methods to plan treatments for circulating tumor cells, disseminated tumor cells, and micrometastases. Given the capacity of radiopharmaceuticals to specifically target and kill single cells and multicellular clusters, a quality not available in chemotherapy and external-beam radiation therapy, it is important to develop dosimetry and bioeffect modeling tools that can inform radiopharmaceutical design and predict their effect on microscopic disease. This pamphlet describes a new version of MIRDcell, a software tool that was initially released by the MIRD committee several years ago. Methods: Version 3 (V3) of MIRDcell uses a combination of analytic and Monte Carlo methods to conduct dosimetry and bioeffect modeling for radiolabeled cells within planar colonies and multicellular clusters. A worked example is provided to assist users to learn old and new features of MIRDcell and test its capacity to recapitulate published responses of tumor cell spheroids to radiopharmaceutical treatments. Prominent capabilities of the new version include radially dependent activity distributions, user-imported activity distributions, cold regions within the cluster, complex bioeffect modeling that accounts for radiation type and subcellular distribution, and a rich table of output data for subsequent analysis. Results: MIRDcell V3 effectively reproduces experimental responses of multicellular spheroids to uniform and nonuniform distributions of therapeutic radiopharmaceuticals. Conclusion: MIRDcell is a versatile software tool that can be used for educational purposes and design of radiopharmaceutical therapies.
Collapse
Affiliation(s)
- Sumudu Katugampola
- Division of Radiation Research, Department of Radiology, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Jianchao Wang
- Division of Radiation Research, Department of Radiology, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Alex Rosen
- Division of Radiation Research, Department of Radiology, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Roger W Howell
- Division of Radiation Research, Department of Radiology, New Jersey Medical School, Rutgers University, Newark, New Jersey
| |
Collapse
|
18
|
Hu B, Liu T, Li L, Shi L, Yao M, Li C, Ma X, Zhu H, Jia B, Wang F. IgG-Binding Nanobody Capable of Prolonging Nanobody-Based Radiotracer Plasma Half-Life and Enhancing the Efficacy of Tumor-Targeted Radionuclide Therapy. Bioconjug Chem 2022; 33:1328-1339. [PMID: 35687724 DOI: 10.1021/acs.bioconjchem.2c00209] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Nanobodies have been developed rapidly as targeted probes for molecular imaging owing to their high affinity, outstanding tissue penetration, and rapid blood clearance. However, the short retention time at the tumor site limits their application in targeted radionuclide therapy. In this study, we designed a dual-targeting nanobody referred to as MIRC213-709, which can specifically bind to the HER2 receptor in tumor cell lines with high affinity (by nanobody MIRC213) and endogenous IgG in plasma to prolong the half-life by the MIRC213 C-terminal fusion nanobody, MIRC709. The nanobodies were site-specifically radiolabeled with 99mTc and 177Lu, and radiochemical purity was >95% after purification. The long blood circulation time and tumor retention property of 99mTc/177Lu-MIRC213-709 were confirmed by a blood clearance assay, single-photon emission computed tomography (SPECT), and a biodistribution study. The blood clearance assay showed that the distribution phase half-life (T1/2α) and elimination phase half-life (T1/2β) of 99mTc-MIRC213-709 were 6.74- and 19.04-fold longer than those of 99mTc-MIRC213, respectively. The SPECT/CT and biodistribution results showed that the highest uptake of 177Lu-MIRC213 in the NCI-N87 model was 5.24 ± 0.95% ID/g at 6 h p.i., while the highest uptake of 177Lu-MIRC213-709 in the NCI-N87 model was 30.82 ± 7.29% ID/g at 48 h p.i. Compared with 177Lu-MIRC213, 177Lu-MIRC213-709 had a 16.9-fold increased tumor cumulative uptake (2606 ± 195.1 vs 153.9 ± 22.37% ID/g·h). The targeted radionuclide therapy assay was performed in the NCI-N87 tumor model, and treatment monitoring ended on day 32. The post-treatment/pretreatment tumor volumes were 12.99 ± 1.66, 3.58 ± 0.96, 1.26 ± 0.17, and 1.54 ± 0.50 in the 0, 9, and 18 MBq single-dose groups and the two 9 MBq divided dose group (14 days apart), respectively. All treatment groups showed significant therapeutic effects (P < 0.0001). Thus, fusion with the IgG-binding nanobody MIRC709 provides MIRC213 derivatives with improved metabolic properties for targeted radionuclide therapy.
Collapse
Affiliation(s)
- Biao Hu
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Tianyu Liu
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Liqiang Li
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Linqing Shi
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Meinan Yao
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Chenzhen Li
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Xiaopan Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Hua Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Bing Jia
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China.,Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Fan Wang
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
19
|
Schneider M, Winkler K, Kell R, Pfaffl MW, Atkinson MJ, Moertl S. The Chaperone Protein GRP78 Promotes Survival and Migration of Head and Neck Cancer After Direct Radiation Exposure and Extracellular Vesicle-Transfer. Front Oncol 2022; 12:842418. [PMID: 35299733 PMCID: PMC8921984 DOI: 10.3389/fonc.2022.842418] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/09/2022] [Indexed: 01/01/2023] Open
Abstract
Background and Purpose Increased levels of the chaperone protein GRP78 have been implicated in poorer outcomes of cancer therapy. We have therefore explored the functional connection between the expression of GRP78 and the development of radioresistance and metastatic behavior in HNSCC. Material and Methods The association between gene expression of GRP78 and survival in HNSCC patients was examined using the TCGA database. The influence of ionizing radiation on the GRP78 levels in HNSCC cell lines, their secreted extracellular vesicles (EV) and non-irradiated EV-recipient cells was investigated by Western Blot and FACS. The consequences of chemical inhibition or experimental overexpression of GRP78 on radioresistance and migration of HNSCC cells were analyzed by clonogenic survival and gap closure assays. Results Elevated levels of GRP78 RNA in HNSCC correlated with poorer overall survival. Radiation increased GRP78 protein expression on the surface of HNSCC cell lines. Experimental overexpression of GRP78 increased both radioresistance and migratory potential. Chemical inhibition of GRP78 impaired cell migration. EVs were identified as a potential source of increased GRP78 content as elevated levels of surface GRP78 were found in EVs released by irradiated cells. These vesicles transferred GRP78 to non-irradiated recipient cells during co-cultivation. Conclusions We have identified the chaperone protein GRP78 as a potential driver of increased radioresistance and motility in HNSCC. The uptake of GRP78-rich EVs originating from irradiated cells may contribute to a poorer prognosis through bystander effects mediated by the transfer of GRP78 to non-irradiated cells. Therefore, we consider the chaperone protein GRP78 to be an attractive target for improving radiotherapy strategies.
Collapse
Affiliation(s)
- Michael Schneider
- Institute of Radiation Medicine, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Klaudia Winkler
- Institute of Radiation Medicine, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Rosemarie Kell
- Institute of Radiation Medicine, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Michael W Pfaffl
- Animal Physiology and Immunology, TUM School of Life Science, Technical University of Munich, Freising, Germany
| | - Michael J Atkinson
- Chair of Radiation Biology, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Simone Moertl
- Department of Effects and Risks of Ionising and Non-Ionising Radiation, Federal Office for Radiation Protection, Oberschleißheim, Germany
| |
Collapse
|
20
|
No Intercellular Regulation of the Cell Cycle among Human Cervical Carcinoma HeLa Cells Expressing Fluorescent Ubiquitination-Based Cell-Cycle Indicators in Modulated Radiation Fields. Int J Mol Sci 2021; 22:ijms222312785. [PMID: 34884589 PMCID: PMC8657989 DOI: 10.3390/ijms222312785] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 11/17/2022] Open
Abstract
The non-targeted effects of radiation have been known to induce significant alternations in cell survival. Although the effects might govern the progression of tumor sites following advanced radiotherapy, the impacts on the intercellular control of the cell cycle following radiation exposure with a modified field, remain to be determined. Recently, a fluorescent ubiquitination-based cell-cycle indicator (FUCCI), which can visualize the cell-cycle phases with fluorescence microscopy in real time, was developed for biological cell research. In this study, we investigated the non-targeted effects on the regulation of the cell cycle of human cervical carcinoma (HeLa) cells with imperfect p53 function that express the FUCCI (HeLa–FUCCI cells). The possible effects on the cell-cycle phases via soluble factors were analyzed following exposure to different field configurations, which were delivered using a 150 kVp X-ray irradiator. In addition, using synchrotron-generated, 5.35 keV monochromatic X-ray microbeams, high-precision 200 μm-slit microbeam irradiation was performed to investigate the possible impacts on the cell-cycle phases via cell–cell contacts. Collectively, we could not detect the intercellular regulation of the cell cycle in HeLa–FUCCI cells, which suggested that the unregulated cell growth was a malignant tumor. Our findings indicated that there was no significant intercellular control system of the cell cycle in malignant tumors during or after radiotherapy, highlighting the differences between normal tissue and tumor characteristics.
Collapse
|
21
|
Kadhim M, Tuncay Cagatay S, Elbakrawy EM. Non-targeted effects of radiation: a personal perspective on the role of exosomes in an evolving paradigm. Int J Radiat Biol 2021; 98:410-420. [PMID: 34662248 DOI: 10.1080/09553002.2021.1980630] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
PURPOSE Radiation-induced non-targeted effects (NTE) have implications in a variety of areas relevant to radiation biology. Here we evaluate the various cargo associated with exosomal signalling and how they work synergistically to initiate and propagate the non-targeted effects including Genomic Instability and Bystander Effects. CONCLUSIONS Extra cellular vesicles, in particular exosomes, have been shown to carry bystander signals. Exosome cargo may contain nucleic acids, both DNA and RNA, as well as proteins, lipids and metabolites. These cargo molecules have all been considered as potential mediators of NTE. A review of current literature shows mounting evidence of a role for ionizing radiation in modulating both the numbers of exosomes released from affected cells as well as the content of their cargo, and that these exosomes can instigate functional changes in recipient cells. However, there are significant gaps in our understanding, particularly regarding modified exosome cargo after radiation exposure and the functional changes induced in recipient cells.
Collapse
Affiliation(s)
- Munira Kadhim
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, United Kingdom
| | - Seda Tuncay Cagatay
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, United Kingdom
| | - Eman Mohammed Elbakrawy
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, United Kingdom.,Department of Radiation Physics, National Center for Radiation Research and Technology, Atomic Energy Authority, 3 Ahmed El-Zomor Al Manteqah Ath Thamenah, Nasr City, Cairo 11787, Egypt
| |
Collapse
|
22
|
Tudor M, Gilbert A, Lepleux C, Temelie M, Hem S, Armengaud J, Brotin E, Haghdoost S, Savu D, Chevalier F. A Proteomic Study Suggests Stress Granules as New Potential Actors in Radiation-Induced Bystander Effects. Int J Mol Sci 2021; 22:ijms22157957. [PMID: 34360718 PMCID: PMC8347418 DOI: 10.3390/ijms22157957] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 01/07/2023] Open
Abstract
Besides the direct effects of radiations, indirect effects are observed within the surrounding non-irradiated area; irradiated cells relay stress signals in this close proximity, inducing the so-called radiation-induced bystander effect. These signals received by neighboring unirradiated cells induce specific responses similar with those of direct irradiated cells. To understand the cellular response of bystander cells, we performed a 2D gel-based proteomic study of the chondrocytes receiving the conditioned medium of low-dose irradiated chondrosarcoma cells. The conditioned medium was directly analyzed by mass spectrometry in order to identify candidate bystander factors involved in the signal transmission. The proteomic analysis of the bystander chondrocytes highlighted 20 proteins spots that were significantly modified at low dose, implicating several cellular mechanisms, such as oxidative stress responses, cellular motility, and exosomes pathways. In addition, the secretomic analysis revealed that the abundance of 40 proteins in the conditioned medium of 0.1 Gy irradiated chondrosarcoma cells was significantly modified, as compared with the conditioned medium of non-irradiated cells. A large cluster of proteins involved in stress granules and several proteins involved in the cellular response to DNA damage stimuli were increased in the 0.1 Gy condition. Several of these candidates and cellular mechanisms were confirmed by functional analysis, such as 8-oxodG quantification, western blot, and wound-healing migration tests. Taken together, these results shed new lights on the complexity of the radiation-induced bystander effects and the large variety of the cellular and molecular mechanisms involved, including the identification of a new potential actor, namely the stress granules.
Collapse
Affiliation(s)
- Mihaela Tudor
- Department of Life and Environmental Physics, HoriaHulubei National Institute of Physics and Nuclear Engineering, 077125 Magurele, Romania; (M.T.); (M.T.); (D.S.)
- Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
| | - Antoine Gilbert
- UMR6252 CIMAP, Team Applications in Radiobiology with Accelerated Ions, CEA-CNRS-ENSICAEN-Université de Caen Normandie, 14000 Caen, France; (A.G.); (C.L.); (S.H.)
| | - Charlotte Lepleux
- UMR6252 CIMAP, Team Applications in Radiobiology with Accelerated Ions, CEA-CNRS-ENSICAEN-Université de Caen Normandie, 14000 Caen, France; (A.G.); (C.L.); (S.H.)
| | - Mihaela Temelie
- Department of Life and Environmental Physics, HoriaHulubei National Institute of Physics and Nuclear Engineering, 077125 Magurele, Romania; (M.T.); (M.T.); (D.S.)
| | - Sonia Hem
- BPMP, Montpellier University, CNRS, INRAE, Institut Agro, 34000 Montpellier, France;
| | - Jean Armengaud
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, 30200 Bagnols-sur-Cèze, France;
| | - Emilie Brotin
- ImpedanCELL Platform, Federative Structure 4206 ICORE, NormandieUniv, UNICAEN, Inserm U1086 ANTICIPE, Biology and Innovative Therapeutics for Ovarian Cancers Group (BioTICLA), Comprehensive Cancer Center F. Baclesse, 14000 Caen, France;
| | - Siamak Haghdoost
- UMR6252 CIMAP, Team Applications in Radiobiology with Accelerated Ions, CEA-CNRS-ENSICAEN-Université de Caen Normandie, 14000 Caen, France; (A.G.); (C.L.); (S.H.)
| | - Diana Savu
- Department of Life and Environmental Physics, HoriaHulubei National Institute of Physics and Nuclear Engineering, 077125 Magurele, Romania; (M.T.); (M.T.); (D.S.)
| | - François Chevalier
- UMR6252 CIMAP, Team Applications in Radiobiology with Accelerated Ions, CEA-CNRS-ENSICAEN-Université de Caen Normandie, 14000 Caen, France; (A.G.); (C.L.); (S.H.)
- Correspondence: ; Tel.: +33-(0)231-454-564
| |
Collapse
|
23
|
Forrester HB, Lobachevsky PN, Stevenson AW, Hall CJ, Martin OA, Sprung CN. Abscopal Gene Expression in Response to Synchrotron Radiation Indicates a Role for Immunological and DNA Damage Response Genes. Radiat Res 2021; 194:678-687. [PMID: 32991732 DOI: 10.1667/rade-19-00014.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 08/24/2020] [Indexed: 11/03/2022]
Abstract
Abscopal effects are an important aspect of targeted radiation therapy due to their implication in normal tissue toxicity from chronic inflammatory responses and mutagenesis. Gene expression can be used to determine abscopal effects at the molecular level. Synchrotron microbeam radiation therapy utilizing high-intensity X rays collimated into planar microbeams is a promising cancer treatment due to its reported ability to ablate tumors with less damage to normal tissues compared to conventional broadbeam radiation therapy techniques. The low scatter of synchrotron radiation enables microbeams to be delivered to tissue effectively, and is also advantageous for out-of-field studies because there is minimal interference from scatter. Mouse legs were irradiated at a dose rate of 49 Gy/s and skin samples in the out-of-field areas were collected. The out-of-field skin showed an increase in Tnf expression and a decrease in Mdm2 expression, genes associated with inflammation and DNA damage. These expression effects from microbeam exposure were similar to those found with broadbeam exposure. In immune-deficient Ccl2 knockout mice, we identified a different gene expression profile which showed an early increase in Mdm2, Tgfb1, Tnf and Ccl22 expression in out-of-field skin that was not observed in the immune-proficient mice. Our results suggest that the innate immune system is involved in out-of-field tissue responses and alterations in the immune response may not eliminate abscopal effects, but could change them.
Collapse
Affiliation(s)
- Helen B Forrester
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia.,Monash University, Clayton, Australia.,School of Science, RMIT University, Melbourne, Australia
| | - Pavel N Lobachevsky
- Research Division, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia.,Advanced Analytical Technologies, Melbourne, Australia
| | - Andrew W Stevenson
- Australian Synchrotron, ANSTO, Clayton, Australia.,CSIRO Manufacturing, Clayton, Australia
| | | | - Olga A Martin
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Carl N Sprung
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia.,Monash University, Clayton, Australia
| |
Collapse
|
24
|
Swati, Chadha VD. Role of epigenetic mechanisms in propagating off-targeted effects following radiation based therapies - A review. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2021; 787:108370. [PMID: 34083045 DOI: 10.1016/j.mrrev.2021.108370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 12/17/2022]
Abstract
Despite being an important diagnostic and treatment modality, ionizing radiation (IR) is also known to cause genotoxicity and multiple side effects leading to secondary carcinogenesis. While modern cancer radiation therapy has improved patient recovery and enhanced survival rates, the risk of radiation-related adverse effects has become a growing challenge. It is now well-accepted that IR-induced side effects are not exclusively restricted to exposed cells but also spread to distant 'bystander' cells and even to the unexposed progeny of the irradiated cells. These 'off-targeted' effects involve a plethora of molecular events depending on the type of radiation and tumor tissue background. While the mechanisms by which off-targeted effects arise remain obscure, emerging evidence based on the non-mendelian inheritance of various manifestations of them as well as their persistence for longer periods supports a contribution of epigenetic factors. This review focuses on the major epigenetic phenomena including DNA methylation, histone modifications, and small RNA mediated silencing and their versatile role in the manifestation of IR induced off-targeted effects. As short- and long-range communication vehicles respectively, the role of gap junctions and exosomes in spreading these epigenetic-alteration driven off-targeted effects is also discussed. Furthermore, this review emphasizes the possible therapeutic potentials of these epigenetic mechanisms and how beneficial outcomes could potentially be achieved by targeting various signaling molecules involved in these mechanisms.
Collapse
Affiliation(s)
- Swati
- Centre for Nuclear Medicine (U.I.E.A.S.T), South Campus, Panjab University, Sector 25, Chandigarh, 160014, India.
| | - Vijayta D Chadha
- Centre for Nuclear Medicine (U.I.E.A.S.T), South Campus, Panjab University, Sector 25, Chandigarh, 160014, India.
| |
Collapse
|
25
|
Cao Y, Xie L, Shi F, Tang M, Li Y, Hu J, Zhao L, Zhao L, Yu X, Luo X, Liao W, Bode AM. Targeting the signaling in Epstein-Barr virus-associated diseases: mechanism, regulation, and clinical study. Signal Transduct Target Ther 2021; 6:15. [PMID: 33436584 PMCID: PMC7801793 DOI: 10.1038/s41392-020-00376-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/30/2020] [Accepted: 10/15/2020] [Indexed: 12/11/2022] Open
Abstract
Epstein–Barr virus-associated diseases are important global health concerns. As a group I carcinogen, EBV accounts for 1.5% of human malignances, including both epithelial- and lymphatic-originated tumors. Moreover, EBV plays an etiological and pathogenic role in a number of non-neoplastic diseases, and is even involved in multiple autoimmune diseases (SADs). In this review, we summarize and discuss some recent exciting discoveries in EBV research area, which including DNA methylation alterations, metabolic reprogramming, the changes of mitochondria and ubiquitin-proteasome system (UPS), oxidative stress and EBV lytic reactivation, variations in non-coding RNA (ncRNA), radiochemotherapy and immunotherapy. Understanding and learning from this advancement will further confirm the far-reaching and future value of therapeutic strategies in EBV-associated diseases.
Collapse
Affiliation(s)
- Ya Cao
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China. .,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China. .,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China. .,Research Center for Technologies of Nucleic Acid-Based Diagnostics and Therapeutics Hunan Province, 410078, Changsha, China. .,Molecular Imaging Research Center of Central South University, 410008, Changsha, Hunan, China. .,National Joint Engineering Research Center for Genetic Diagnostics of Infectious Diseases and Cancer, 410078, Changsha, China. .,Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.
| | - Longlong Xie
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China
| | - Feng Shi
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China
| | - Min Tang
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China.,Molecular Imaging Research Center of Central South University, 410008, Changsha, Hunan, China
| | - Yueshuo Li
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China
| | - Jianmin Hu
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China
| | - Lin Zhao
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China
| | - Luqing Zhao
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China
| | - Xinfang Yu
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China
| | - Xiangjian Luo
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China.,Molecular Imaging Research Center of Central South University, 410008, Changsha, Hunan, China
| | - Weihua Liao
- Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| |
Collapse
|
26
|
The role of connexin proteins and their channels in radiation-induced atherosclerosis. Cell Mol Life Sci 2021; 78:3087-3103. [PMID: 33388835 PMCID: PMC8038956 DOI: 10.1007/s00018-020-03716-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/29/2020] [Accepted: 11/17/2020] [Indexed: 02/08/2023]
Abstract
Radiotherapy is an effective treatment for breast cancer and other thoracic tumors. However, while high-energy radiotherapy treatment successfully kills cancer cells, radiation exposure of the heart and large arteries cannot always be avoided, resulting in secondary cardiovascular disease in cancer survivors. Radiation-induced changes in the cardiac vasculature may thereby lead to coronary artery atherosclerosis, which is a major cardiovascular complication nowadays in thoracic radiotherapy-treated patients. The underlying biological and molecular mechanisms of radiation-induced atherosclerosis are complex and still not fully understood, resulting in potentially improper radiation protection. Ionizing radiation (IR) exposure may damage the vascular endothelium by inducing DNA damage, oxidative stress, premature cellular senescence, cell death and inflammation, which act to promote the atherosclerotic process. Intercellular communication mediated by connexin (Cx)-based gap junctions and hemichannels may modulate IR-induced responses and thereby the atherosclerotic process. However, the role of endothelial Cxs and their channels in atherosclerotic development after IR exposure is still poorly defined. A better understanding of the underlying biological pathways involved in secondary cardiovascular toxicity after radiotherapy would facilitate the development of effective strategies that prevent or mitigate these adverse effects. Here, we review the possible roles of intercellular Cx driven signaling and communication in radiation-induced atherosclerosis.
Collapse
|
27
|
Belli M, Indovina L. The Response of Living Organisms to Low Radiation Environment and Its Implications in Radiation Protection. Front Public Health 2020; 8:601711. [PMID: 33384980 PMCID: PMC7770185 DOI: 10.3389/fpubh.2020.601711] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022] Open
Abstract
Life has evolved on Earth for about 4 billion years in the presence of the natural background of ionizing radiation. It is extremely likely that it contributed, and still contributes, to shaping present form of life. Today the natural background radiation is extremely small (few mSv/y), however it may be significant enough for living organisms to respond to it, perhaps keeping memory of this exposure. A better understanding of this response is relevant not only for improving our knowledge on life evolution, but also for assessing the robustness of the present radiation protection system at low doses, such as those typically encountered in everyday life. Given the large uncertainties in epidemiological data below 100 mSv, quantitative evaluation of these health risk is currently obtained with the aid of radiobiological models. These predict a health detriment, caused by radiation-induced genetic mutations, linearly related to the dose. However a number of studies challenged this paradigm by demonstrating the occurrence of non-linear responses at low doses, and of radioinduced epigenetic effects, i.e., heritable changes in genes expression not related to changes in DNA sequence. This review is focused on the role that epigenetic mechanisms, besides the genetic ones, can have in the responses to low dose and protracted exposures, particularly to natural background radiation. Many lines of evidence show that epigenetic modifications are involved in non-linear responses relevant to low doses, such as non-targeted effects and adaptive response, and that genetic and epigenetic effects share, in part, a common origin: the reactive oxygen species generated by ionizing radiation. Cell response to low doses of ionizing radiation appears more complex than that assumed for radiation protection purposes and that it is not always detrimental. Experiments conducted in underground laboratories with very low background radiation have even suggested positive effects of this background. Studying the changes occurring in various living organisms at reduced radiation background, besides giving information on the life evolution, have opened a new avenue to answer whether low doses are detrimental or beneficial, and to understand the relevance of radiobiological results to radiation protection.
Collapse
Affiliation(s)
| | - Luca Indovina
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
28
|
Howell RW. Advancements in the use of Auger electrons in science and medicine during the period 2015-2019. Int J Radiat Biol 2020; 99:2-27. [PMID: 33021416 PMCID: PMC8062591 DOI: 10.1080/09553002.2020.1831706] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/01/2020] [Accepted: 09/28/2020] [Indexed: 02/06/2023]
Abstract
Auger electrons can be highly radiotoxic when they are used to irradiate specific molecular sites. This has spurred basic science investigations of their radiobiological effects and clinical investigations of their potential for therapy. Focused symposia on the biophysical aspects of Auger processes have been held quadrennially. This 9th International Symposium on Physical, Molecular, Cellular, and Medical Aspects of Auger Processes at Oxford University brought together scientists from many different fields to review past findings, discuss the latest studies, and plot the future work to be done. This review article examines the research in this field that was published during the years 2015-2019 which corresponds to the period since the last meeting in Japan. In addition, this article points to future work yet to be done. There have been a plethora of advancements in our understanding of Auger processes. These advancements range from basic atomic and molecular physics to new ways to implement Auger electron emitters in radiopharmaceutical therapy. The highly localized doses of radiation that are deposited within a 10 nm of the decay site make them precision tools for discovery across the physical, chemical, biological, and medical sciences.
Collapse
Affiliation(s)
- Roger W Howell
- Division of Radiation Research, Department of Radiology, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| |
Collapse
|
29
|
Pakniyat F, Nedaie HA, Mozdarani H, Mahmoudzadeh A, Salimi M, Griffin RJ, Gholami S. Enhanced response of radioresistant carcinoma cell line to heterogeneous dose distribution of grid; the role of high-dose bystander effect. Int J Radiat Biol 2020; 96:1585-1596. [PMID: 33074047 DOI: 10.1080/09553002.2020.1834163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE The classical dogma that restricted the radiation effect to the directly irradiated cells has been challenged by the bystander effect. This off-target phenomenon which was manifested in adjacent cells via signaling of fully exposed cells might be involved in high-dose Grid therapy as well. Here, an in-vitro study was performed to examine the possible extent of carcinoma cells response to the inhomogeneous dose distribution of Grid irradiation in the context of the bystander effect. MATERIALS AND METHODS Bystander effect was investigated in human carcinoma cell lines of HeLa and HN5 adjacent to those received high-dose Grid irradiation using 'medium transfer' and 'cell-to-cell contact' strategies. Based on the Grid peak-to-valley dose profile, medium transfer was exerted from 10 Gy uniformly exposed donors to 1.5 Gy uniformly irradiated recipients. Cell-contact bystander was evaluated after nonuniform dose distribution of 10 Gy Grid irradiation using cloning cylinders. GammaH2AX foci, micronucleus and clonogenic assays besides gene expression analysis were performed. RESULTS Various parameters (ɑ/β, D37, D50) extracted from survival curve which fitted to the Linear Quadratic model, verified more radioresistance of HN5. Survival fraction at 2 Gy (SF2) indicated as 0.42 ± 0.06 in HeLa and 0.5 ± 0.03 in HN5. The level of survival decrease, DNA damages and micronucleus of cells located in the Grid shielded areas (1.5 Gy cell-to-cell contact bystander cells) were significantly more than the values obtained from cells which were irradiated by merely uniform dose of 1.5 Gy. The gH2AX foci and micronuclei frequencies were enhanced in cell-contact bystander approximately more than 1.8 times. Relative expression of DNA damage repair pathway genes (Xrcc6 and H2afx) in bystander cells increased significantly. The most cell survival reduction (11.6 times) was revealed in the Grid bystander cells of radioresistant cell line (HN5). No statistically significant difference between 10 Gy uniform beam and Grid non-uniform beam was observed. CONCLUSIONS Various endpoints confirmed an augmented response of cells in the valley dose region of the Grid block significantly (compared with the cells irradiated by identical dose of uniform beam), suggesting the role of high-dose bystander effect which was more pronounced in resistant carcinoma cell lines. These findings could provide a partial explanation for the Grid beneficial response seen in a number of pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Fatemeh Pakniyat
- Department of Medical Physics and Biomedical Engineering, Tehran University of Medical Sciences, Tehran, Iran
| | - Hassan Ali Nedaie
- Department of Medical Physics and Biomedical Engineering, Tehran University of Medical Sciences, Tehran, Iran.,Radiation Oncology Research Center, Cancer institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Mozdarani
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Aziz Mahmoudzadeh
- Department of Bioscience and Biotechnology, Malek-Ashtar University of Technology, Tehran, Iran
| | - Mahdieh Salimi
- Department of Medical genetics, Medical Biotechnology Institute, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Robert J Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Somayeh Gholami
- Radiation Oncology Research Center, Cancer institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Pourhajibagher M, Ahmadi H, Roshan Z, Bahador A. Streptococcus mutans bystander-induced bioeffects following sonodynamic antimicrobial chemotherapy through sonocatalytic performance of Curcumin-Poly (Lactic-co-Glycolic Acid) on off-target cells. Photodiagnosis Photodyn Ther 2020; 32:102022. [PMID: 33038486 DOI: 10.1016/j.pdpdt.2020.102022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/01/2020] [Accepted: 09/18/2020] [Indexed: 02/06/2023]
Abstract
To assessed the Streptococcus mutans bystander-induced bioeffects following sonodynamic antimicrobial chemotherapy (SACT) by Curcumin-Poly (Lactic-co-Glycolic Acid) nanoparticles (Cur-PLGA-NPs). Cur-PLGA-NPs were synthesized and characterized by Scanning Electron Microscope (SEM), Transmission Electron Microscope (TEM), and Attenuated Total Reflection Fourier Transform IR (ATR-FTIR) spectroscopy, as well as, determination of in vitro drug release. Following the successful synthesis and characterization of Cur-PLGA-NPs, the cell survival, intracellular ROS production, apoptotic effects, DNA fragmentation, and gene expression levels of pro-inflammatory cytokines were investigated on human gingival fibroblast (HGF) cells as off-target cells through S. mutans bystander-induced bioeffects following SACT (BCSS). No significant cytotoxic and damage caused by the release of ROS from BCSS were observed in HGF cells (P > 0.05). There was no DNA fragmentation and anti-proliferation effects on HGF cells. The expression levels of bFGF, TNF-α, and IL-8 genes were increased after exposure to BCSS to 15.4-, 13.5-, and 8.7-fold, respectively (P < 0.05), while TGF-ß and IL-10 were downregulated to -4.1- and -6.8-fold, respectively (P < 0.05). It could be concluded that there were no bystander bioeffects of targeted sonocatalytic stress on off-target cells.
Collapse
Affiliation(s)
- Maryam Pourhajibagher
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hanie Ahmadi
- Department of Polymer Engineering and Color Technology, Amirkabir University of Technology, Tehran, Iran
| | - Zahra Roshan
- Department of Polymer Engineering and Color Technology, Amirkabir University of Technology, Tehran, Iran
| | - Abbas Bahador
- Oral Microbiology Laboratory, Department of Medical Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Fellowship in Clinical Laboratory Sciences, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
31
|
Ionizing Radiation-Induced Epigenetic Modifications and Their Relevance to Radiation Protection. Int J Mol Sci 2020; 21:ijms21175993. [PMID: 32825382 PMCID: PMC7503247 DOI: 10.3390/ijms21175993] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022] Open
Abstract
The present system of radiation protection assumes that exposure at low doses and/or low dose-rates leads to health risks linearly related to the dose. They are evaluated by a combination of epidemiological data and radiobiological models. The latter imply that radiation induces deleterious effects via genetic mutation caused by DNA damage with a linear dose-dependence. This picture is challenged by the observation of radiation-induced epigenetic effects (changes in gene expression without altering the DNA sequence) and of non-linear responses, such as non-targeted and adaptive responses, that in turn can be controlled by gene expression networks. Here, we review important aspects of the biological response to ionizing radiation in which epigenetic mechanisms are, or could be, involved, focusing on the possible implications to the low dose issue in radiation protection. We examine in particular radiation-induced cancer, non-cancer diseases and transgenerational (hereditary) effects. We conclude that more realistic models of radiation-induced cancer should include epigenetic contribution, particularly in the initiation and progression phases, while the impact on hereditary risk evaluation is expected to be low. Epigenetic effects are also relevant in the dispute about possible "beneficial" effects at low dose and/or low dose-rate exposures, including those given by the natural background radiation.
Collapse
|
32
|
Pellizzon ACA. Lattice radiation therapy - its concept and impact in the immunomodulation cancer treatment era. ACTA ACUST UNITED AC 2020; 66:728-731. [PMID: 32696876 DOI: 10.1590/1806-9282.66.6.728] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 12/08/2019] [Indexed: 04/05/2023]
Abstract
Voluminous tumors represent a challenge in radiation oncology, particularly when surgical resection is not possible. Lattice radiotherapy (LTR) is a technique that may provide equivalent or superior clinical response in the management of large tumors while limiting toxicity to adjacent normal tissues. LRT can precisely deliver inhomogeneous high doses of radiation to different areas within the gross tumor volumes (GTV). The dosimetric characteristic of LTR is defined by the ratio of the valley dose (lower doses - cold spots) and the peak doses, also called vertex (higher doses - hot spots), or the valley-to-peak dose ratio. The valley-to-peak ratio thereby quantifies the degree of spatial fractionation. LRT delivers high doses of radiation without exceeding the tolerance of adjacent critical structures. Radiobiological experiments support the role of radiation-induced bystander effects, vascular alterations, and immunologic interactions in areas subject to low dose radiation. The technological advancements continue to expand in Radiation Oncology, bringing new safety opportunities of treatment for bulky lesions.
Collapse
|
33
|
Radiation-induced bystander and abscopal effects: important lessons from preclinical models. Br J Cancer 2020; 123:339-348. [PMID: 32581341 PMCID: PMC7403362 DOI: 10.1038/s41416-020-0942-3] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 03/10/2020] [Accepted: 05/28/2020] [Indexed: 12/11/2022] Open
Abstract
Radiotherapy is a pivotal component in the curative treatment of patients with localised cancer and isolated metastasis, as well as being used as a palliative strategy for patients with disseminated disease. The clinical efficacy of radiotherapy has traditionally been attributed to the local effects of ionising radiation, which induces cell death by directly and indirectly inducing DNA damage, but substantial work has uncovered an unexpected and dual relationship between tumour irradiation and the host immune system. In clinical practice, it is, therefore, tempting to tailor immunotherapies with radiotherapy in order to synergise innate and adaptive immunity against cancer cells, as well as to bypass immune tolerance and exhaustion, with the aim of facilitating tumour regression. However, our understanding of how radiation impacts on immune system activation is still in its early stages, and concerns and challenges regarding therapeutic applications still need to be overcome. With the increasing use of immunotherapy and its common combination with ionising radiation, this review briefly delineates current knowledge about the non-targeted effects of radiotherapy, and aims to provide insights, at the preclinical level, into the mechanisms that are involved with the potential to yield clinically relevant combinatorial approaches of radiotherapy and immunotherapy.
Collapse
|
34
|
Hamis S, Powathil GG, Chaplain MAJ. Blackboard to Bedside: A Mathematical Modeling Bottom-Up Approach Toward Personalized Cancer Treatments. JCO Clin Cancer Inform 2020; 3:1-11. [PMID: 30742485 DOI: 10.1200/cci.18.00068] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cancers present with high variability across patients and tumors; thus, cancer care, in terms of disease prevention, detection, and control, can highly benefit from a personalized approach. For a comprehensive personalized oncology practice, this personalization should ideally consider data gathered from various information levels, which range from the macroscale population level down to the microscale tumor level, without omission of the central patient level. Appropriate data mined from each of these levels can significantly contribute in devising personalized treatment plans tailored to the individual patient and tumor. Mathematical models of solid tumors, combined with patient-specific tumor profiles, present a unique opportunity to personalize cancer treatments after detection using a bottom-up approach. Here, we discuss how information harvested from mathematical models and from corresponding in silico experiments can be implemented in preclinical and clinical applications. To conceptually illustrate the power of these models, one such model is presented, and various pertinent tumor and treatment scenarios are demonstrated in silico. The presented model, specifically a multiscale, hybrid cellular automaton, has been fully validated in vitro using multiple cell-line-specific data. We discuss various insights provided by this model and other models like it and their role in designing predictive tools that are both patient, and tumor specific. After refinement and parametrization with appropriate data, such in silico tools have the potential to be used in a clinical setting to aid in treatment protocols and decision making.
Collapse
Affiliation(s)
- Sara Hamis
- Swansea University, Swansea, Wales, United Kingdom
| | | | | |
Collapse
|
35
|
Brito AE, Etchebehere E. Radium-223 as an Approved Modality for Treatment of Bone Metastases. Semin Nucl Med 2020; 50:177-192. [PMID: 32172803 DOI: 10.1053/j.semnuclmed.2019.11.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Radium-223 dichloride (223Ra) is an α-emitter radionuclide approved for treatment of osteoblastic metastases in castrate-resistant prostate cancer (mCRPC) patients. 223Ra increases overall survival, improves bone pain, increases the median time to the first skeletal-related event, reduces the use of external beam radiation therapy for bone pain palliation, reduces the rates of spinal cord compression, and hospitalization. 223Ra therapy has minimal side effects; the most common hematological side effects are anemia, thrombocytopenia and neutropenia while the nonhematological side effects that may occur are bone pain flare, nausea, fatigue, and diarrhea. Alongside 223Ra therapy there are currently a variety of first-line therapeutic options available to treat mCRPC patients and much debate regarding the appropriate treatment algorithm for these patients and the possible combination of therapies among the ones available. In this article, we review the rationale behind 223Ra therapy as well as 223Ra mechanisms of action, biodistribution and dosimetry, optimal timing possibilities to initiate 223Ra in contrast to other treatments available, the association of 223Ra with other therapies and the means of evaluating patients in order to properly deliver to 223Ra therapy. Furthermore, we will discuss 223Ra dose administration possibilities, patient and dose preparation and the challenges of treatment response evaluation during and after 223Ra.
Collapse
Affiliation(s)
- Ana Emília Brito
- Real Nuclear, Real Hospital Português de Beneficência em Pernambuco, Recife, Brazil
| | - Elba Etchebehere
- Division of Nuclear Medicine, The University of Campinas (UNICAMP), Campinas, Brazil.
| |
Collapse
|
36
|
Arbabi Moghadam S, Rezania V, Tuszynski JA. Cell death and survival due to cytotoxic exposure modelled as a two-state Ising system. ROYAL SOCIETY OPEN SCIENCE 2020; 7:191578. [PMID: 32257323 PMCID: PMC7062046 DOI: 10.1098/rsos.191578] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 01/08/2020] [Indexed: 06/11/2023]
Abstract
Cancer chemotherapy agents are assessed for their therapeutic utility primarily by their ability to cause apoptosis of cancer cells and their potency is given by an IC50 value. Chemotherapy uses both target-specific and systemic-action drugs and drug combinations to treat cancer. It is important to judiciously choose a drug type, its dosage and schedule for optimized drug selection and administration. Consequently, the precise mathematical formulation of cancer cells' response to chemotherapy may assist in the selection process. In this paper, we propose a mathematical description of the cancer cell response to chemotherapeutic agent exposure based on a time-tested physical model of two-state multiple-component systems near criticality. We describe the Ising model methodology and apply it to a diverse panel of cytotoxic drugs administered against numerous cancer cell lines in a dose-response manner. The analysed dataset was generated by the Netherlands Translational Research Center B.V. (Oncolines). This approach allows for an accurate and consistent analysis of cytotoxic agents' effects on cancer cell lines and reveals the presence or absence of the bystander effect through the interaction constant. By calculating the susceptibility function, we see the value of IC50 coinciding with the peak of this measure of the system's sensitivity to external perturbations.
Collapse
Affiliation(s)
- S. Arbabi Moghadam
- Department of Physics, University of Alberta, Edmonton, Alberta, Canada T6G 2E1
| | - V. Rezania
- Department of Physical Sciences, MacEwan University, Edmonton, Alberta, Canada T5 J 4S2
| | - J. A. Tuszynski
- Department of Physics, University of Alberta, Edmonton, Alberta, Canada T6G 2E1
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada T6G 1Z2
- DIMEAS, Politecnico di Torino, Turin, Italy
| |
Collapse
|
37
|
Thariat J, Valable S, Laurent C, Haghdoost S, Pérès EA, Bernaudin M, Sichel F, Lesueur P, Césaire M, Petit E, Ferré AE, Saintigny Y, Skog S, Tudor M, Gérard M, Thureau S, Habrand JL, Balosso J, Chevalier F. Hadrontherapy Interactions in Molecular and Cellular Biology. Int J Mol Sci 2019; 21:E133. [PMID: 31878191 PMCID: PMC6981652 DOI: 10.3390/ijms21010133] [Citation(s) in RCA: 12] [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: 11/22/2019] [Revised: 12/17/2019] [Accepted: 12/20/2019] [Indexed: 02/06/2023] Open
Abstract
The resistance of cancer cells to radiotherapy is a major issue in the curative treatment of cancer patients. This resistance can be intrinsic or acquired after irradiation and has various definitions, depending on the endpoint that is chosen in assessing the response to radiation. This phenomenon might be strengthened by the radiosensitivity of surrounding healthy tissues. Sensitive organs near the tumor that is to be treated can be affected by direct irradiation or experience nontargeted reactions, leading to early or late effects that disrupt the quality of life of patients. For several decades, new modalities of irradiation that involve accelerated particles have been available, such as proton therapy and carbon therapy, raising the possibility of specifically targeting the tumor volume. The goal of this review is to examine the up-to-date radiobiological and clinical aspects of hadrontherapy, a discipline that is maturing, with promising applications. We first describe the physical and biological advantages of particles and their application in cancer treatment. The contribution of the microenvironment and surrounding healthy tissues to tumor radioresistance is then discussed, in relation to imaging and accurate visualization of potentially resistant hypoxic areas using dedicated markers, to identify patients and tumors that could benefit from hadrontherapy over conventional irradiation. Finally, we consider combined treatment strategies to improve the particle therapy of radioresistant cancers.
Collapse
Affiliation(s)
- Juliette Thariat
- Department of Radiation Oncology, Centre François Baclesse, 14000 Caen, France; (J.T.); (P.L.); (M.C.); (M.G.); (J.-L.H.); (J.B.)
- Laboratoire de Physique Corpusculaire IN2P3/ENSICAEN-UMR6534-Unicaen-Normandie Université, 14000 Caen, France;
- ARCHADE Research Community, 14000 Caen, France; (S.V.); (C.L.); (S.H.); (E.A.P.); (M.B.); (F.S.); (E.P.); (A.E.F.); (Y.S.)
| | - Samuel Valable
- ARCHADE Research Community, 14000 Caen, France; (S.V.); (C.L.); (S.H.); (E.A.P.); (M.B.); (F.S.); (E.P.); (A.E.F.); (Y.S.)
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, 14000 Caen, France
| | - Carine Laurent
- ARCHADE Research Community, 14000 Caen, France; (S.V.); (C.L.); (S.H.); (E.A.P.); (M.B.); (F.S.); (E.P.); (A.E.F.); (Y.S.)
- Normandie Univ, UNICAEN, UNIROUEN, ABTE, 14000 Caen, France
| | - Siamak Haghdoost
- ARCHADE Research Community, 14000 Caen, France; (S.V.); (C.L.); (S.H.); (E.A.P.); (M.B.); (F.S.); (E.P.); (A.E.F.); (Y.S.)
- LARIA, iRCM, François Jacob Institute, DRF-CEA, 14000 Caen, France
- UMR6252 CIMAP, CEA-CNRS-ENSICAEN-Université de Caen Normandie, 14000 Caen, France;
| | - Elodie A. Pérès
- ARCHADE Research Community, 14000 Caen, France; (S.V.); (C.L.); (S.H.); (E.A.P.); (M.B.); (F.S.); (E.P.); (A.E.F.); (Y.S.)
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, 14000 Caen, France
| | - Myriam Bernaudin
- ARCHADE Research Community, 14000 Caen, France; (S.V.); (C.L.); (S.H.); (E.A.P.); (M.B.); (F.S.); (E.P.); (A.E.F.); (Y.S.)
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, 14000 Caen, France
| | - François Sichel
- ARCHADE Research Community, 14000 Caen, France; (S.V.); (C.L.); (S.H.); (E.A.P.); (M.B.); (F.S.); (E.P.); (A.E.F.); (Y.S.)
- Normandie Univ, UNICAEN, UNIROUEN, ABTE, 14000 Caen, France
| | - Paul Lesueur
- Department of Radiation Oncology, Centre François Baclesse, 14000 Caen, France; (J.T.); (P.L.); (M.C.); (M.G.); (J.-L.H.); (J.B.)
- ARCHADE Research Community, 14000 Caen, France; (S.V.); (C.L.); (S.H.); (E.A.P.); (M.B.); (F.S.); (E.P.); (A.E.F.); (Y.S.)
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, 14000 Caen, France
| | - Mathieu Césaire
- Department of Radiation Oncology, Centre François Baclesse, 14000 Caen, France; (J.T.); (P.L.); (M.C.); (M.G.); (J.-L.H.); (J.B.)
- ARCHADE Research Community, 14000 Caen, France; (S.V.); (C.L.); (S.H.); (E.A.P.); (M.B.); (F.S.); (E.P.); (A.E.F.); (Y.S.)
| | - Edwige Petit
- ARCHADE Research Community, 14000 Caen, France; (S.V.); (C.L.); (S.H.); (E.A.P.); (M.B.); (F.S.); (E.P.); (A.E.F.); (Y.S.)
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, 14000 Caen, France
| | - Aurélie E. Ferré
- ARCHADE Research Community, 14000 Caen, France; (S.V.); (C.L.); (S.H.); (E.A.P.); (M.B.); (F.S.); (E.P.); (A.E.F.); (Y.S.)
- Normandie Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP CYCERON, 14000 Caen, France
| | - Yannick Saintigny
- ARCHADE Research Community, 14000 Caen, France; (S.V.); (C.L.); (S.H.); (E.A.P.); (M.B.); (F.S.); (E.P.); (A.E.F.); (Y.S.)
- LARIA, iRCM, François Jacob Institute, DRF-CEA, 14000 Caen, France
- UMR6252 CIMAP, CEA-CNRS-ENSICAEN-Université de Caen Normandie, 14000 Caen, France;
| | - Sven Skog
- Sino-Swed Molecular Bio-Medicine Research Institute, Shenzhen 518057, China;
| | - Mihaela Tudor
- UMR6252 CIMAP, CEA-CNRS-ENSICAEN-Université de Caen Normandie, 14000 Caen, France;
- Department of Life and Environmental Physics, Horia Hulubei National Institute of Physics and Nuclear Engineering, PO Box MG-63, 077125 Magurele, Romania
- Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, R-050095 Bucharest, Romania
| | - Michael Gérard
- Department of Radiation Oncology, Centre François Baclesse, 14000 Caen, France; (J.T.); (P.L.); (M.C.); (M.G.); (J.-L.H.); (J.B.)
- ARCHADE Research Community, 14000 Caen, France; (S.V.); (C.L.); (S.H.); (E.A.P.); (M.B.); (F.S.); (E.P.); (A.E.F.); (Y.S.)
| | - Sebastien Thureau
- Laboratoire de Physique Corpusculaire IN2P3/ENSICAEN-UMR6534-Unicaen-Normandie Université, 14000 Caen, France;
- Department of Radiation Oncology, Centre Henri Becquerel, 76000 Rouen, France
| | - Jean-Louis Habrand
- Department of Radiation Oncology, Centre François Baclesse, 14000 Caen, France; (J.T.); (P.L.); (M.C.); (M.G.); (J.-L.H.); (J.B.)
- ARCHADE Research Community, 14000 Caen, France; (S.V.); (C.L.); (S.H.); (E.A.P.); (M.B.); (F.S.); (E.P.); (A.E.F.); (Y.S.)
- Normandie Univ, UNICAEN, UNIROUEN, ABTE, 14000 Caen, France
| | - Jacques Balosso
- Department of Radiation Oncology, Centre François Baclesse, 14000 Caen, France; (J.T.); (P.L.); (M.C.); (M.G.); (J.-L.H.); (J.B.)
- ARCHADE Research Community, 14000 Caen, France; (S.V.); (C.L.); (S.H.); (E.A.P.); (M.B.); (F.S.); (E.P.); (A.E.F.); (Y.S.)
| | - François Chevalier
- ARCHADE Research Community, 14000 Caen, France; (S.V.); (C.L.); (S.H.); (E.A.P.); (M.B.); (F.S.); (E.P.); (A.E.F.); (Y.S.)
- LARIA, iRCM, François Jacob Institute, DRF-CEA, 14000 Caen, France
- UMR6252 CIMAP, CEA-CNRS-ENSICAEN-Université de Caen Normandie, 14000 Caen, France;
| |
Collapse
|
38
|
Rodríguez-Barbeito P, Díaz-Botana P, Gago-Arias A, Feijoo M, Neira S, Guiu-Souto J, López-Pouso Ó, Gómez-Caamaño A, Pardo-Montero J. A Model of Indirect Cell Death Caused by Tumor Vascular Damage after High-Dose Radiotherapy. Cancer Res 2019; 79:6044-6053. [PMID: 31641030 DOI: 10.1158/0008-5472.can-19-0181] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 07/02/2019] [Accepted: 10/16/2019] [Indexed: 11/16/2022]
Abstract
There is increasing evidence that high doses of radiotherapy, like those delivered in stereotactic body radiotherapy (SBRT), trigger indirect mechanisms of cell death. Such effect seems to be two-fold. High doses may trigger an immune response and may cause vascular damage, leading to cell starvation and death. Development of mathematical response models, including indirect death, may help clinicians to design SBRT optimal schedules. Despite increasing experimental literature on indirect tumor cell death caused by vascular damage, efforts on modeling this effect have been limited. In this work, we present a biomathematical model of this effect. In our model, tumor oxygenation is obtained by solving the reaction-diffusion equation; radiotherapy kills tumor cells according to the linear-quadratic model, and also endothelial cells (EC), which can trigger loss of functionality of capillaries. Capillary death will affect tumor oxygenation, driving nearby tumor cells into severe hypoxia. Capillaries can recover functionality due to EC proliferation. Tumor cells entering a predetermined severe hypoxia status die according to a hypoxia-death model. This model fits recently published experimental data showing the effect of vascular damage on surviving fractions. It fits surviving fraction curves and qualitatively reproduces experimental values of percentages of functional capillaries 48 hours postirradiation, and hypoxic cells pre- and 48 hours postirradiation. This model is useful for exploring aspects of tumor and EC response to radiotherapy and constitutes a stepping stone toward modeling indirect tumor cell death caused by vascular damage and accounting for this effect during SBRT planning. SIGNIFICANCE: A novel biomathematical model of indirect tumor cell death caused by vascular radiation damage could potentially help clinicians interpret experimental data and design better radiotherapy schedules.
Collapse
Affiliation(s)
- Pedro Rodríguez-Barbeito
- Group of Medical Physics and Biomathematics, Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain.,Department of Applied Mathematics, Universidade de Santiago de Compostela, Spain
| | - Pablo Díaz-Botana
- Group of Medical Physics and Biomathematics, Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain.,Galician Supercomputation Center (CESGA), Santiago de Compostela, Spain
| | - Araceli Gago-Arias
- Group of Medical Physics and Biomathematics, Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain.,Institute of Physics, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
| | - Manuel Feijoo
- Department of Particle Physics, Universidade de Santiago de Compostela, Spain
| | - Sara Neira
- Group of Medical Physics and Biomathematics, Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
| | - Jacobo Guiu-Souto
- Department of Medical Physics, Complexo Hospitalario Universitario de Santiago de Compostela, Spain.,Department of Medical Physics, Fundación Centro Oncolóxico de Galicia, A Coruña, Spain
| | - Óscar López-Pouso
- Group of Medical Physics and Biomathematics, Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain.,Department of Applied Mathematics, Universidade de Santiago de Compostela, Spain
| | - Antonio Gómez-Caamaño
- Department of Radiotherapy, Complexo Hospitalario Universitario de Santiago de Compostela, Spain
| | - Juan Pardo-Montero
- Group of Medical Physics and Biomathematics, Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain. .,Department of Medical Physics, Complexo Hospitalario Universitario de Santiago de Compostela, Spain
| |
Collapse
|
39
|
Heeran AB, Berrigan HP, O'Sullivan J. The Radiation-Induced Bystander Effect (RIBE) and its Connections with the Hallmarks of Cancer. Radiat Res 2019; 192:668-679. [PMID: 31618121 DOI: 10.1667/rr15489.1] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Radiation therapy is one of the pillars of cancer treatment, with approximately one half of all cancer patients receiving it as part of their standard of care. Emerging evidence indicates that the biological effects of radiation are not limited to targeted cells. The radiation-induced bystander effect (RIBE) refers to the plethora of biological phenomena occurring in nonirradiated cells as a result of signal transmission from an irradiated cell. Experimental evidence has linked RIBE to numerous hallmarks of cancer including resisting cell death, tumor immune evasion, genomic instability, deregulated cellular energetics, tumor-promoting inflammation and sustained proliferative signaling as well as enhanced radioresistance, thus highlighting the potential role of RIBE events in patient treatment response. The mechanisms underlying RIBE events in vivo are poorly understood. However, elucidating the molecular mechanisms involved in their manifestation may reveal novel therapeutic targets to improve radiation response in cancer patients.
Collapse
Affiliation(s)
- Aisling B Heeran
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James's Hospital, Dublin 8, Ireland
| | - Helen P Berrigan
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James's Hospital, Dublin 8, Ireland
| | - Jacintha O'Sullivan
- Trinity Translational Medicine Institute, Department of Surgery, Trinity College Dublin and St. James's Hospital, Dublin 8, Ireland
| |
Collapse
|
40
|
De Marzi L, Nauraye C, Lansonneur P, Pouzoulet F, Patriarca A, Schneider T, Guardiola C, Mammar H, Dendale R, Prezado Y. Spatial fractionation of the dose in proton therapy: Proton minibeam radiation therapy. Cancer Radiother 2019; 23:677-681. [DOI: 10.1016/j.canrad.2019.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 08/01/2019] [Indexed: 10/26/2022]
|
41
|
Kanagaraj K, Rajan V, Pandey BN, Thayalan K, Venkatachalam P. Primary and secondary bystander effect and genomic instability in cells exposed to high and low linear energy transfer radiations. Int J Radiat Biol 2019; 95:1648-1658. [PMID: 31486717 DOI: 10.1080/09553002.2019.1665208] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Purpose: Non-Targeted effects (NTE), such as bystander effect (BE) and genomic instability (GI) challenge central dogma of radiation biology. Moreover, there is a need to understand its universality in different type of cells and radiation quality.Materials and method: To study BE (primary and secondary) and GI Human adult dermal fibroblast (HADF) and peripheral blood lymphocytes (PBL) were exposed to low fluence of 241Am alpha (α) particle and 6 MV X-ray. The BE was carried out by means of co-culture methodology after exposing the cells to both types of radiation and damage was measured using micronucleus assay (MN) and chromosomal aberration assay (CA) in the p1 cells while the GI was followed up in their progeny.Results: A dose-dependent increase in DNA damages (MN and CA) was observed in directly irradiated and bystander cells. The magnitude of BE was higher (6 fold) in cells co-cultured with the α-irradiated cells than that of with X-irradiated cells. Cross exposure of both cell types confirms that radiation induced BE is cell type dependent. In addition, induced DNA damage persisted for a longer population doubling in α-particle irradiated cells.Conclusion: This work adds evidence to secondary bystander response generated from primary bystander normal cells and its dependence to radiation quality.
Collapse
Affiliation(s)
- K Kanagaraj
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | - V Rajan
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Badri N Pandey
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - K Thayalan
- Department of Radiation oncology, Kamakshi Memorial Hospital, Chennai, India
| | - P Venkatachalam
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| |
Collapse
|
42
|
Tumor-derived extracellular vesicles: insights into bystander effects of exosomes after irradiation. Lasers Med Sci 2019; 35:531-545. [PMID: 31529349 DOI: 10.1007/s10103-019-02880-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 09/06/2019] [Indexed: 12/20/2022]
Abstract
This review article aims to address the kinetic of TDEs in cancer cells pre- and post-radiotherapy. Radiotherapy is traditionally used for the treatment of multiple cancer types; however, there is growing evidence to show that radiotherapy exerts NTEs on cells near to the irradiated cells. In tumor mass, irradiated cells can affect non-irradiated cells in different ways. Of note, exosomes are nano-scaled cell particles releasing from tumor cells and play key roles in survival, metastasis, and immunosuppression of tumor cells. Recent evidence indicated that irradiation has the potential to affect the dynamic of different signaling pathways such as exosome biogenesis. Indeed, exosomes act as intercellular mediators in various cell communication through transmitting bio-molecules. Due to their critical roles in cancer biology, exosomes are at the center of attention. TDEs contain an exclusive molecular signature that they may serve as tumor biomarker in the diagnosis of different cancers. Interestingly, radiotherapy and IR could also contribute to altering the dynamic of exosome secretion. Most probably, the content of exosomes in irradiated cells is different compared to exosomes originated from the non-irradiated BCs. Irradiated cells release exosomes with exclusive content that mediate NTEs in BCs. Considering variation in cell type, IR doses, and radio-resistance or radio-sensitivity of different cancers, there is, however, contradictions in the feature and activity of irradiated exosomes on neighboring cells.
Collapse
|
43
|
Leung CN, Canter BS, Rajon D, Bäck TA, Fritton JC, Azzam EI, Howell RW. Dose-Dependent Growth Delay of Breast Cancer Xenografts in the Bone Marrow of Mice Treated with 223Ra: The Role of Bystander Effects and Their Potential for Therapy. J Nucl Med 2019; 61:89-95. [PMID: 31519805 DOI: 10.2967/jnumed.119.227835] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 06/07/2019] [Indexed: 01/12/2023] Open
Abstract
The role of radiation-induced bystander effects in radiation therapy remains unclear. With renewed interest in therapy with α-particle emitters, and their potential for sterilizing disseminated tumor cells (DTCs), it is critical to determine the contribution of bystander effects to the overall response so they can be leveraged for maximum clinical benefit. Methods: Female Foxn1nu athymic nude mice were administered 0, 50, or 600 kBq/kg 223RaCl2 to create bystander conditions. At 24 hours after administration, MDA-MB-231 or MCF-7 human breast cancer cells expressing luciferase were injected into the tibial marrow compartment. Tumor burden was tracked weekly via bioluminescence. Results: The MDA-MB-231 xenografts were observed to have a 10-day growth delay in the 600 kBq/kg treatment group only. In contrast, MCF-7 cells had 7- and 65-day growth delays in the 50 and 600 kBq/kg groups, respectively. Histologic imaging of the tibial marrow compartment, α-camera imaging, and Monte Carlo dosimetry modeling revealed DTCs both within and beyond the range of the α-particles emitted from 223Ra in bone for both MCF-7 and MDA-MB-231 cells. Conclusion: Taken together, these results support the participation of 223Ra-induced antiproliferative/cytotoxic bystander effects in delayed growth of DTC xenografts. They indicate that the delay depends on the injected activity and therefore is dose-dependent. They suggest using 223RaCl2 as an adjuvant treatment for select patients at early stages of breast cancer.
Collapse
Affiliation(s)
- Calvin N Leung
- Department of Radiology, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Brian S Canter
- Department of Radiology, New Jersey Medical School, Rutgers University, Newark, New Jersey.,Department of Orthopedics, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Didier Rajon
- Department of Neurosurgery, University of Florida, Gainesville, Florida; and
| | - Tom A Bäck
- Department of Radiation Physics, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - J Christopher Fritton
- Department of Orthopedics, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Edouard I Azzam
- Department of Radiology, New Jersey Medical School, Rutgers University, Newark, New Jersey
| | - Roger W Howell
- Department of Radiology, New Jersey Medical School, Rutgers University, Newark, New Jersey
| |
Collapse
|
44
|
Billena C, Khan AJ. A Current Review of Spatial Fractionation: Back to the Future? Int J Radiat Oncol Biol Phys 2019; 104:177-187. [PMID: 30684666 PMCID: PMC7443362 DOI: 10.1016/j.ijrobp.2019.01.073] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 12/13/2018] [Accepted: 01/15/2019] [Indexed: 11/24/2022]
Abstract
Spatially fractionated radiation therapy represents a significant departure from canonical thinking in radiation oncology despite having origins in the early 1900s. The original and most common implementation of spatially fractionated radiation therapy uses commercially available blocks or multileaf collimators to deliver a nonconfluent, sieve-like pattern of radiation to the target volume in a nonuniform dose distribution. Dosimetrically, this is parameterized by the ratio of the valley dose in cold spots to the peak dose in hot spots, or the valley-to-peak dose ratio. The radiobiologic mechanisms are postulated to involve radiation-induced bystander effects, microvascular alterations, and/or immunomodulation. Current indications include bulky or locally advanced disease that would not be amenable to conventional radiation or that has proved refractory to chemoradiation. Early-phase clinical trials have shown remarkable success, with some response rates >90% and minimal toxicity. This has promoted technological developments in 3-dimensional formats (LATTICE), micron-size beams (microbeam), and proton arrays. Nevertheless, more clinical and biological data are needed to specify ideal dosimetry parameters and to formulate robust clinical indications and guidelines for optimal standardized care.
Collapse
Affiliation(s)
- Cole Billena
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Atif J Khan
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
45
|
Changes of microbial cell survival, metabolic activity, efflux capacity, and quorum sensing ability of Aggregatibacter actinomycetemcomitans due to antimicrobial photodynamic therapy-induced bystander effects. Photodiagnosis Photodyn Ther 2019; 26:287-294. [PMID: 31026616 DOI: 10.1016/j.pdpdt.2019.04.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 04/22/2019] [Accepted: 04/22/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND The bystander effects, whereby naive (bystander) microbial cells near microbial cells directly exposed to certain treatment show responses that would not have happened in the absence of the directly targeted microbial cells, is recently documented in the field of microbiology. In this article, we discuss that substantial bystander responses are also observed after antimicrobial photodynamic therapy (aPDT) using curcumin (Cur). MATERIALS AND METHODS Bystander effects induced by whole bacterial cell suspension (WBCST), cell-free supernatants fluid (CFSFT), and bacterial cell pellet (BCPT) obtained from A. actinomycetemcomitans culture treated with Cur-aPDT on cell survival, quorum sensing (QS) ability, metabolic activity and efflux capacity of A. actinomycetemcomitans were determined using microbial viability assay, Escherichia coli-based bioassay, XTT reduction method, and ethidium bromide (EtBr) accumulation assay, respectively. RESULTS A. actinomycetemcomitans cell survival reduced by 82.7% (P = 0.001) and 76.2% (P = 0.01) after exposure to WBCST and CFSFT, respectively. The A. actinomycetemcomitans population increased by 5.5% (P = 0.7) after exposure to BCPT. Bacterial metabolic activity decreased by 42.6% (P = 0.02), 35.3% (P = 0.03), and 9.4% (P = 0.5) after exposure to WBCST, CFSFT, and BCPT, respectively. A. actinomycetemcomitans exposed to WBCST, CFSFT, and BCPT showed a reduction of 83.2% (P = 0.001), 77.2% (P = 0.01) and 21.9% (P = 0.09) in the QS mediator compared to the WBCSU, CFSFU, and BCPU of untreated A. actinomycetemcomitans, respectively. No significant change of the EtBr accumulation was observed in the three preparations of the Cur-aPDT-treated culture (i.e. WBCST, CFSFT, and BCPT) compared to their respective controls. CONCLUSIONS The results of the current study revealed that Cur-aPDT could significantly reduce microbial cell survival, cell metabolic activity, efflux capacity, and QS ability through the bystander effects. As a result, the bystander effects of Cur-aPDT along with the direct effect of Cur-aPDT can enhance the efficiency of aPDT as an adjunct therapeutic strategy for treatment of local infections.
Collapse
|
46
|
Fukunaga H, Yokoya A, Taki Y, Butterworth KT, Prise KM. Precision Radiotherapy and Radiation Risk Assessment: How Do We Overcome Radiogenomic Diversity? TOHOKU J EXP MED 2019; 247:223-235. [PMID: 30971620 DOI: 10.1620/tjem.247.223] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Precision medicine is a rapidly developing area that aims to deliver targeted therapies based on individual patient characteristics. However, current radiation treatment is not yet personalized; consequently, there is a critical need for specific patient characteristics of both tumor and normal tissues to be fully incorporated into dose prescription. Furthermore, current risk assessment following environmental, occupational, or accidental exposures to radiation is based on population effects, and does not account for individual diversity underpinning radiosensitivity. The lack of personalized approaches in both radiotherapy and radiation risk assessment resulted in the current situation where a population-based model, effective dose, is being used. In this review article, to stimulate scientific discussion for precision medicine in both radiotherapy and radiation risk assessment, we propose a novel radiological concept and metric - the personalized dose and the personalized risk index - that incorporate individual physiological, lifestyle-related and genomic variations and radiosensitivity, outlining the potential clinical application for precision medicine. We also review on recent progress in both genomics and biobanking research, which is promising for providing novel insights into individual radiosensitivity, and for creating a novel conceptual framework of precision radiotherapy and radiation risk assessment.
Collapse
Affiliation(s)
- Hisanori Fukunaga
- Centre for Cancer Research and Cell Biology, Queen's University Belfast
| | - Akinari Yokoya
- Tokai Quantum Beam Science Center, National Institutes for Quantum and Radiological Science and Technology
| | - Yasuyuki Taki
- Institute of Development, Aging and Cancer, Tohoku University
| | | | - Kevin M Prise
- Centre for Cancer Research and Cell Biology, Queen's University Belfast
| |
Collapse
|
47
|
Bystander effectors of chondrosarcoma cells irradiated at different LET impair proliferation of chondrocytes. J Cell Commun Signal 2019; 13:343-356. [PMID: 30903603 PMCID: PMC6732157 DOI: 10.1007/s12079-019-00515-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 03/08/2019] [Indexed: 12/17/2022] Open
Abstract
While the dose-response relationship of radiation-induced bystander effect (RIBE) is controversial at low and high linear energy transfer (LET), mechanisms and effectors of cell-to-cell communication stay unclear and highly dependent of cell type. In the present study, we investigated the capacity of chondrocytes in responding to bystander factors released by chondrosarcoma cells irradiated at different doses (0.05 to 8 Gy) with X-rays and C-ions. Following a medium transfer protocol, cell survival, proliferation and DNA damages were quantified in bystander chondrocytes. The bystander factors secreted by chondrosarcoma cells were characterized. A significant and major RIBE response was observed in chondrocyte cells (T/C-28a2) receiving conditioned medium from chondrosarcoma cells (SW1353) irradiated with 0.1 Gy of X-rays and 0.05 Gy of C-ions, resulting in cell survivals of 36% and 62%, respectively. Micronuclei induction in bystander cells was observed from the same low doses. The cell survival results obtained by clonogenic assays were confirmed using impedancemetry. The bystander activity was vanished after a heat treatment or a dilution of the conditioned media. The cytokines which are well known as bystander factors, TNF-α and IL-6, were increased as a function of doses and LET according to an ELISA multiplex analysis. Together, the results demonstrate that irradiated chondrosarcoma cells can communicate stress factors to non-irradiated chondrocytes, inducing a wide and specific bystander response related to both doses and LET.
Collapse
|
48
|
Evolution of the Supermodel: Progress in Modelling Radiotherapy Response in Mice. Clin Oncol (R Coll Radiol) 2019; 31:272-282. [PMID: 30871751 DOI: 10.1016/j.clon.2019.02.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 02/14/2019] [Accepted: 02/15/2019] [Indexed: 12/18/2022]
Abstract
Mouse models are essential tools in cancer research that have been used to understand the genetic basis of tumorigenesis, cancer progression and to test the efficacies of anticancer treatments including radiotherapy. They have played a critical role in our understanding of radiotherapy response in tumours and normal tissues and continue to evolve to better recapitulate the underlying biology of humans. In addition, recent developments in small animal irradiators have significantly improved in vivo irradiation techniques, allowing previously unimaginable experimental approaches to be explored in the laboratory. The combination of contemporary mouse models with small animal irradiators represents a major step forward for the radiobiology field in being able to much more accurately replicate clinical exposure scenarios. As radiobiology studies become ever more sophisticated in reflecting developments in the clinic, it is increasingly important to understand the basis and potential limitations of extrapolating data from mice to humans. This review provides an overview of mouse models and small animal radiotherapy platforms currently being used as advanced radiobiological research tools towards improving the translational power of preclinical studies.
Collapse
|
49
|
Farhood B, Goradel NH, Mortezaee K, Khanlarkhani N, Salehi E, Nashtaei MS, Shabeeb D, Musa AE, Fallah H, Najafi M. Intercellular communications-redox interactions in radiation toxicity; potential targets for radiation mitigation. J Cell Commun Signal 2019; 13:3-16. [PMID: 29911259 PMCID: PMC6381372 DOI: 10.1007/s12079-018-0473-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 06/12/2018] [Indexed: 12/20/2022] Open
Abstract
Nowadays, using ionizing radiation (IR) is necessary for clinical, agricultural, nuclear energy or industrial applications. Accidental exposure to IR after a radiation terror or disaster poses a threat to human. In contrast to the old dogma of radiation toxicity, several experiments during the last two recent decades have revealed that intercellular signaling and communications play a key role in this procedure. Elevated level of cytokines and other intercellular signals increase oxidative damage and inflammatory responses via reduction/oxidation interactions (redox system). Intercellular signals induce production of free radicals and inflammatory mediators by some intermediate enzymes such as cyclooxygenase-2 (COX-2), nitric oxide synthase (NOS), NADPH oxidase, and also via triggering mitochondrial ROS. Furthermore, these signals facilitate cell to cell contact and increasing cell toxicity via cohort effect. Nitric oxide is a free radical with ability to act as an intercellular signal that induce DNA damage and changes in some signaling pathways in irradiated as well as non-irradiated adjacent cells. Targeting of these mediators by some anti-inflammatory agents or via antioxidants such as mitochondrial ROS scavengers opens a window to mitigate radiation toxicity after an accidental exposure. Experiments which have been done so far suggests that some cytokines such as IL-1β, TNF-α, TGF-β, IL-4 and IL-13 are some interesting targets that depend on irradiated organs and may help mitigate radiation toxicity. Moreover, animal experiments in recent years indicated that targeting of toll like receptors (TLRs) may be more useful for radioprotection and mitigation. In this review, we aimed to describe the role of intercellular interactions in oxidative injury, inflammation, cell death and killing effects of IR. Moreover, we described evidence on potential mitigation of radiation injury via targeting of these mediators.
Collapse
Affiliation(s)
- Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Nasser Hashemi Goradel
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Neda Khanlarkhani
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ensieh Salehi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Shabani Nashtaei
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Infertility Department, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Dheyauldeen Shabeeb
- Department of Medical Physics & Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, International Campus, Tehran, Iran
- Department of Physiology, College of Medicine, University of Misan, Misan, Iraq
| | - Ahmed Eleojo Musa
- Department of Medical Physics & Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, International Campus, Tehran, Iran
- Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran
| | - Hengameh Fallah
- Department of Chemistry, Faculty of Science, Islamic Azad University, Arak, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
50
|
Ji K, Wang Y, Du L, Xu C, Liu Y, He N, Wang J, Liu Q. Research Progress on the Biological Effects of Low-Dose Radiation in China. Dose Response 2019; 17:1559325819833488. [PMID: 30833876 PMCID: PMC6393828 DOI: 10.1177/1559325819833488] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/19/2018] [Accepted: 12/29/2018] [Indexed: 01/07/2023] Open
Abstract
Human are exposed to ionizing radiation from natural and artificial sources, which consequently poses a possible risk to human health. However, accumulating evidence indicates that the biological effects of low-dose radiation (LDR) are different from those of high-dose radiation (HDR). Low-dose radiation–induced hormesis has been extensively observed in different biological systems, including immunological and hematopoietic systems. Adaptive responses in response to LDR that can induce cellular resistance to genotoxic effects from subsequent exposure to HDR have also been described and researched. Bystander effects, another type of biological effect induced by LDR, have been shown to widely occur in many cell types. Furthermore, the influence of LDR-induced biological effects on certain diseases, such as cancer and diabetes, has also attracted the interest of researchers. Many studies have suggested that LDR has the potential antitumor and antidiabetic complications effects. In addition, the researches on whether LDR could induce stochastic effects were also debated. Studies on the biological effects of LDR in China started in 1970s and considerable progress has been made since. In the present article, we provide an overview of the research progress on the biological effects of LDR in China.
Collapse
Affiliation(s)
- Kaihua Ji
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| | - Yan Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| | - Liqing Du
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| | - Chang Xu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| | - Yang Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| | - Ningning He
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| | - Jinhan Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| | - Qiang Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Department of Radiobiology, Institute of Radiation Medicine of Chinese Academy of Medical Science & Pecking Union Medical College, Tianjin, PR China
| |
Collapse
|