1
|
Ruysscher DD, Wauters E, Jendrossek V, Filippi AR, Revel MP, Faivre-Finn C, Naidoo J, Ramella S, Guckenberger M, Ricardi U, Khalil A, Schor M, Bartolomeo V, Putora PM. Diagnosis and treatment of radiation induced pneumonitis in patients with lung cancer: An ESTRO clinical practice guideline. Radiother Oncol 2025; 207:110837. [PMID: 40185160 DOI: 10.1016/j.radonc.2025.110837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 03/01/2025] [Indexed: 04/07/2025]
Abstract
The incidence of radiation pneumonitis (RP) has decreased significantly compared to historical series, mainly due to improved radiotherapy techniques and patient selection. Nevertheless, some patients still develop RP. This guideline provides user-friendly flowcharts to address common clinical practice questions regarding RP. We summarize the current state of the art regarding the mechanisms, risk factors, diagnosis and treatment of RP. Dosimetric constraints to minimize the incidence of RP, as well as risk factors for developing RP, such as idiopathic pulmonary fibrosis (IPF) were identified. The combination of radiotherapy and medication as a risk factor for the development of RP was reviewed. RP remains a diagnosis of exclusion, but an algorithm for reaching the diagnosis has been proposed. Finally, practical approaches to the treatment of RP are outlined.
Collapse
Affiliation(s)
- Dirk De Ruysscher
- Department of Radiation Oncology (Maastro), Maastricht University Medical Centre(+), GROW School for Oncology and Reproduction, Maastricht, the Netherlands; Department of Radiation Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.
| | - Els Wauters
- Department of Respiratory Diseases, Respiratory Oncology Unit, University Hospital KU Leuven, Leuven, Belgium
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, University Hospital Essen, West German Cancer Center Essen, Essen, Germany
| | - Andrea Riccardo Filippi
- Department of Oncology, University of Milan, Milan, Italy; Radiation Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marie-Pierre Revel
- Université Paris Cité, Paris 75006, France; Department of Radiology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris 75014, France
| | - Corinne Faivre-Finn
- Radiotherapy Related Research, University of Manchester and The Christie NHS Foundation, Manchester, UK
| | - Jarushka Naidoo
- Beaumont Hospital and RCSI University of Health Sciences, Dublin, Ireland; Sidney Kimmel Comprehensive Cancer Centre at Johns Hopkins University, Baltimore, USA
| | - Sara Ramella
- Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland
| | | | - Umberto Ricardi
- Department of Oncology, Radiation Oncology, University of Turin, Turin, Italy
| | - Azza Khalil
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Marieke Schor
- UB Education, Content & Support, Maastricht University, Maastricht 6211 LK, the Netherlands
| | - Valentina Bartolomeo
- Department of Radiation Oncology (Maastro), Maastricht University Medical Centre(+), GROW School for Oncology and Reproduction, Maastricht, the Netherlands; Department of Radiation Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands; Radiation Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; Department of Clinical Surgical, Diagnostic and Pediatric Sciences, Pavia University, 27100 Pavia, Italy
| | - Paul Martin Putora
- Department of Radiation Oncology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland; Department of Radiation Oncology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| |
Collapse
|
2
|
Khateeb S, Hassan AI. Insights into nanostructured lipid carriers of etoricoxib for mitigating radiation-induced lung inflammation and exploring anti-inflammatory mechanisms in rats. Fundam Clin Pharmacol 2025; 39:e70014. [PMID: 40325971 DOI: 10.1111/fcp.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 04/03/2025] [Accepted: 04/16/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND Radiation exposure can cause inflammation, which etoricoxib (ET), an anti-inflammatory drug, could potentially mitigate. OBJECTIVE This study aimed to evaluate the potential effectiveness of etoricoxib-loaded nanostructured lipid carriers (ET-NLCs) in mitigating radiation-induced acute lung inflammation in rats. METHODS Thirty-six rats were divided into six groups. Group 1 (C): control; group 2 (ET): normal rats given ET (10 mg/kg) orally for 14 days; group 3 (ET-NLC): normal rats administered ET-NLCs orally (10 mg/kg) for 14 days. Group 4 (R): rats exposed to 6 Gy whole-body gamma radiation, untreated thereafter to induce lung inflammation and injury. Group 5 (ET-R), irradiated rats, were administered ET (10 mg/kg) orally daily for 14 days. Group 6 (ET-NLC-R), irradiated rats, were administered ET-NLCs (10 mg/kg) orally daily for 14 days. Molecular, biochemical, and histopathological analyses were performed to assess inflammation, apoptosis, oxidative stress, and lung tissue architecture. RESULTS Radiation exposure led to a 1053% increase in Bax expression and an 81.5% decrease in Bcl-2, indicating heightened apoptosis. ET-NLCs treatment reversed these effects, reducing Bax by 59.9% and increasing Bcl-2 by 337.4%. Additionally, ET-NLCs reduced caspase-3 and caspase-8 activation by 54.5% and 62.9%, respectively, compared to radiation exposure alone. Furthermore, ET-NLCs demonstrated potent anti-inflammatory effects by reducing interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α) levels by 49% and 39%, respectively, compared to the irradiated group. Radiation increased malondialdehyde (MDA) levels by 388%, indicating oxidative damage, and suppressed antioxidant enzymes such as catalase (CAT), glutathione peroxidase (GPX), and superoxide dismutase (SOD). ET-NLC treatment decreased MDA levels and increased CAT, GPX, and SOD by 35.7%, 4766.7%, and 765.9%, respectively, restoring antioxidant balance. Radiation reduced surfactant protein (SP-D) levels to 4.9% of control values, but ET-NLCs treatment restored them to 14%. Histopathological analysis revealed that radiation-exposed lungs showed thickened inter-alveolar septa, emphysematous areas, and inflammatory infiltration. ET-NLCs treatment exhibited only mild thickening and limited inflammatory cell infiltration, suggesting significant improvement in lung architecture. CONCLUSIONS Based on these results, NLCs are one of the most promising ways to deliver anti-inflammatory drugs for treating lung damage caused by radiation.
Collapse
Affiliation(s)
- Sahar Khateeb
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk, 71491, Saudi Arabia
- Biochemistry Division, Department of Chemistry, Faculty of Science, Fayoum University, Fayoum, Egypt
| | - Amal I Hassan
- Radioisotopes Department, Nuclear Research Centre, Egyptian Atomic Energy Authority, Giza, Egypt
| |
Collapse
|
3
|
Rajabinasab F, Hajimirzaei P, Ramezani F, Moayer F, Gorjipour F, Nikoofar A, Hassanzadeh L, Hamblin MR, Janzadeh A, Paydar R. Apigenin's Influence on Inflammatory and Epigenetic Responses in Rat Lungs After Radiotherapy. Curr Radiopharm 2025; 18:147-157. [PMID: 39444181 DOI: 10.2174/0118744710336823241011095632] [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: 06/11/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION The lung is a moderately radio-sensitive organ. When cells are damaged due to accidental radiation exposure or treatment, they release molecules that lead to the recruitment of immune cells, accumulating inflammatory cytokines at the site of damage. Apigenin (Api) is a natural flavonoid known for its anti-inflammatory properties. In this study, we investigated the radioprotective properties of Api in the lung. METHODS Thirty-six Wistar rats were randomly assigned to nine groups: control, radiation (Rad), CMC+Rad, Api10+Rad, and Api20+Rad. Api was administered with an intraperitoneal injection for 7 days, after which the rats were irradiated with 6 Gy whole-body X-ray. At 6 and 72 hours post-irradiation, the rats were euthanized, and their lung tissue was extracted. RESULTS Radiation led to increased alveolar wall thickness and the infiltration of macrophages and lymphocytes. Furthermore, the expression levels of inflammatory factors such as a nuclear factor of kappa light polypeptide gene enhancer in B-cells (NF-κB), Glycogen synthase kinase-3 beta (GSK-3β), transforming growth factor-beta1 (TGF-β1), and epigenetic factors including DNA methyltransferase 3a (DNMT3a) and Histone deacetylase 2 (HDAC2) were elevated in the lung tissue following radiation. Meanwhile, the expression level of IκB-α decreased. However, administration of Api (at both 10&20 mg/kg) reversed the adverse effects of radiation. CONCLUSION Api administration mitigated radiation-induced lung damage by reversing inflammatory and epigenetic changes.
Collapse
Affiliation(s)
- Fatemeh Rajabinasab
- Department of Radiation Sciences, Allied Medicine Faculty, Iran University of Medical Sciences, Tehran, Iran
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Pooya Hajimirzaei
- Department of Radiation Sciences, Allied Medicine Faculty, Iran University of Medical Sciences, Tehran, Iran
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ramezani
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fariborz Moayer
- Department of Pathobiology, Faculty of Veterinary Medicine, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Fazel Gorjipour
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Nikoofar
- Department of Radiation Oncology, Iran University of Medical Sciences, Tehran, Iran
| | - Leila Hassanzadeh
- Department of Nuclear Medicine, School of Medicine, Rajaie Cardiovascular, Medical & Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Imaging, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Michael R Hamblin
- Laser Research Centre, University of Johannesburg, Doornfontein, South Africa
| | - Atousa Janzadeh
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Paydar
- Department of Radiation Sciences, Allied Medicine Faculty, Iran University of Medical Sciences, Tehran, Iran
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Bertho A, Ortiz R, Maurin M, Juchaux M, Gilbert C, Espenon J, Ramasamy G, Patriarca A, De Marzi L, Pouzoulet F, Prezado Y. Thoracic Proton Minibeam Radiation Therapy: Tissue Preservation and Survival Advantage Over Conventional Proton Therapy. Int J Radiat Oncol Biol Phys 2024; 120:579-592. [PMID: 38621606 DOI: 10.1016/j.ijrobp.2024.04.011] [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: 01/10/2024] [Revised: 03/25/2024] [Accepted: 04/05/2024] [Indexed: 04/17/2024]
Abstract
PURPOSE Proton minibeam radiation therapy (pMBRT) is an innovative radiation therapy approach that highly modulates the spatial dimension of the dose delivery using narrow, parallel, and submillimetric proton beamlets. pMBRT has proven its remarkable healthy tissue preservation in the brain and skin. This study assesses the potential advantages of pMBRT for thoracic irradiations compared with conventional radiation therapy in terms of normal tissue toxicity. The challenge here was the influence of respiratory motion on the typical peak and valley dose patterns of pMBRT and its potential biologic effect. METHODS AND MATERIALS The whole thorax of naïve C57BL/6 mice received one fraction of high dose (18 Gy) pMBRT or conventional proton therapy (CPT) without any respiratory control. The development of radiation-induced pulmonary fibrosis was longitudinally monitored using cone beam computed tomography. Anatomopathologic analysis was carried out at 9 months postirradiation and focused on the reaction of the lungs' parenchyma and the response of cell types involved in the development of radiation-induced fibrosis and lung regeneration as alveolar type II epithelial cells, club cells, and macrophages. RESULTS pMBRT has milder effects on survival, skin reactions, and lung fibrosis compared with CPT. The pMBRT-induced lung changes were more regional and less severe, with evidence of potential reactive proliferation of alveolar type II epithelial cells and less extensive depletion of club cells and macrophage invasion than the more damaging effects observed in CPT. CONCLUSIONS pMBRT appears suitable to treat moving targets, holding a significant ability to preserve healthy lung tissue, even without respiratory control or precise targeting.
Collapse
Affiliation(s)
- Annaïg Bertho
- Institut Curie, Université PSL, CNRS UMR3347, INSERM U1021, Signalisation Radiobiologie et Cancer, Orsay, France; Université Paris-Saclay, CNRS UMR3347, INSERM U1021, Signalisation Radiobiologie et Cancer, Orsay, France
| | - Ramon Ortiz
- Institut Curie, Université PSL, CNRS UMR3347, INSERM U1021, Signalisation Radiobiologie et Cancer, Orsay, France; Université Paris-Saclay, CNRS UMR3347, INSERM U1021, Signalisation Radiobiologie et Cancer, Orsay, France
| | - Mathieu Maurin
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Marjorie Juchaux
- Institut Curie, Université PSL, CNRS UMR3347, INSERM U1021, Signalisation Radiobiologie et Cancer, Orsay, France; Université Paris-Saclay, CNRS UMR3347, INSERM U1021, Signalisation Radiobiologie et Cancer, Orsay, France
| | - Cristèle Gilbert
- Institut Curie, Université PSL, CNRS UMR3347, INSERM U1021, Signalisation Radiobiologie et Cancer, Orsay, France; Université Paris-Saclay, CNRS UMR3347, INSERM U1021, Signalisation Radiobiologie et Cancer, Orsay, France
| | - Julie Espenon
- Institut Curie, Université PSL, CNRS UMR3347, INSERM U1021, Signalisation Radiobiologie et Cancer, Orsay, France; Université Paris-Saclay, CNRS UMR3347, INSERM U1021, Signalisation Radiobiologie et Cancer, Orsay, France
| | - Gabriel Ramasamy
- Institut Curie, PSL Research University, Département de Recherche Translationnelle, CurieCoreTech-Experimental Radiation therapy (RadeXp), Paris, France
| | - Annalisa Patriarca
- Centre de Protonthérapie d'Orsay, Radiation Oncology Department, Campus Universitaire, Institut Curie, PSL University, Orsay, France
| | - Ludovic De Marzi
- Centre de Protonthérapie d'Orsay, Radiation Oncology Department, Campus Universitaire, Institut Curie, PSL University, Orsay, France; Institut Curie, Campus Universitaire, PSL University, University Paris Saclay, INSERM, Orsay
| | - Frédéric Pouzoulet
- Institut Curie, PSL Research University, Département de Recherche Translationnelle, CurieCoreTech-Experimental Radiation therapy (RadeXp), Paris, France; Institut Curie, PSL University, Université Paris-Saclay, Inserm, Laboratoire de Recherche Translationnelle en Oncologie, Orsay, France
| | - Yolanda Prezado
- Institut Curie, Université PSL, CNRS UMR3347, INSERM U1021, Signalisation Radiobiologie et Cancer, Orsay, France; Université Paris-Saclay, CNRS UMR3347, INSERM U1021, Signalisation Radiobiologie et Cancer, Orsay, France.
| |
Collapse
|
5
|
Groves AM, Paris ND, Johnston CJ, Hernady E, Finkelstein J, Lawrence P, Marples B. Mitigating Viral Impact on the Radiation Response of the Lung. Radiat Res 2024; 202:552-564. [PMID: 39048109 PMCID: PMC11610374 DOI: 10.1667/rade-24-00103.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/11/2024] [Indexed: 07/27/2024]
Abstract
Inflammation is a key factor in both influenza and radiation-induced lung pathophysiology. This implies a commonality of response to pulmonary damage from these insults and suggests exacerbated pathology may occur after combined exposure. We therefore tested the hypothesis that past inflammation from viral infection alters the lung microenvironment and lowers tolerance for radiation injury. Mice were inoculated with influenza A virus (IAV) and three weeks later, after virus clearance, mice received total-body irradiation (TBI). Survival as well as systemic and local lung inflammation were assessed, and strategies to mitigate pulmonary injury were investigated. After IAV infection alone, body condition recovered within 3 weeks, however inflammatory pathways remained active for 15 weeks. IAV infection exacerbated subsequent TBI responses, evident by increased lethality, enhanced histologically evident lung injury and an altered lung macrophage phenotype. To mitigate this enhanced sensitivity, captopril [an angiotensin converting enzyme inhibitor (ACEi)] was administered to limit tissue inflammation, or inflammatory monocyte-derived macrophage recruitment was blocked with a C-C chemokine receptor type 2 (CCR2) inhibitor. Both treatments abrogated the changes in circulating immune cells observed 4 weeks after TBI, and attenuated pro-inflammatory phenotypes in lung alveolar macrophages, appearing to shift immune cell dynamics towards recovery. Histologically apparent lung injury was not improved by either treatment. We show that latent lung injury from viral infection exacerbates radiation morbidity and mortality. Although strategies that attenuate proinflammatory immune cell phenotypes can normalize macrophage dynamics, this does not fully mitigate lung injury. Recognizing that past viral infections can enhance lung radiosensitivity is of critical importance for patients receiving TBI, as it could increase the incidence of adverse outcomes.
Collapse
Affiliation(s)
- Angela M. Groves
- Department of Radiation Oncology, University of Rochester, Rochester, New York
| | - Nicole D. Paris
- Department of Radiation Oncology, University of Rochester, Rochester, New York
| | - Carl J. Johnston
- Department of Pediatrics, University of Rochester, Rochester, New York
| | - Eric Hernady
- Department of Radiation Oncology, University of Rochester, Rochester, New York
| | - Jacob Finkelstein
- Department of Pediatrics, University of Rochester, Rochester, New York
| | - Paige Lawrence
- Department of Environmental Medicine, University of Rochester, Rochester, New York
| | - Brian Marples
- Department of Radiation Oncology, University of Rochester, Rochester, New York
| |
Collapse
|
6
|
Blakely WF, Port M, Ostheim P, Abend M. Radiation Research Society Journal-based Historical Review of the Use of Biomarkers for Radiation Dose and Injury Assessment: Acute Health Effects Predictions. Radiat Res 2024; 202:185-204. [PMID: 38936821 DOI: 10.1667/rade-24-00121.1] [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: 05/02/2024] [Accepted: 06/11/2024] [Indexed: 06/29/2024]
Abstract
A multiple-parameter based approach using radiation-induced clinical signs and symptoms, hematology changes, cytogenetic chromosomal aberrations, and molecular biomarkers changes after radiation exposure is used for biodosimetry-based dose assessment. In the current article, relevant milestones from Radiation Research are documented that forms the basis of the current consensus approach for diagnostics after radiation exposure. For example, in 1962 the use of cytogenetic chromosomal aberration using the lymphocyte metaphase spread dicentric assay for biodosimetry applications was first published in Radiation Research. This assay is now complimented using other cytogenetic chromosomal aberration assays (i.e., chromosomal translocations, cytokinesis-blocked micronuclei, premature chromosome condensation, γ-H2AX foci, etc.). Changes in blood cell counts represent an early-phase biomarker for radiation exposures. Molecular biomarker changes have evolved to include panels of organ-specific plasma proteomic and blood-based gene expression biomarkers for radiation dose assessment. Maturation of these assays are shown by efforts for automated processing and scoring, development of point-of-care diagnostics devices, service laboratories inter-comparison exercises, and applications for dose and injury assessments in radiation accidents. An alternative and complementary approach has been advocated with the focus to de-emphasize "dose" and instead focus on predicting acute or delayed health effects. The same biomarkers used for dose estimation (e.g., lymphocyte counts) can be used to directly predict the later developing severity degree of acute health effects without performing dose estimation as an additional or intermediate step. This review illustrates contributing steps toward these developments published in Radiation Research.
Collapse
Affiliation(s)
- William F Blakely
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Matthias Port
- Bundeswehr Institute of Radiobiology, Munich, Germany
| | | | - Michael Abend
- Bundeswehr Institute of Radiobiology, Munich, Germany
| |
Collapse
|
7
|
Zhang Y, Zhou Y, Zhou Y, Yu X, Shen X, Hong Y, Zhang Y, Wang S, Mou M, Zhang J, Tao L, Gao J, Qiu Y, Chen Y, Zhu F. TheMarker: a comprehensive database of therapeutic biomarkers. Nucleic Acids Res 2024; 52:D1450-D1464. [PMID: 37850638 PMCID: PMC10767989 DOI: 10.1093/nar/gkad862] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/21/2023] [Accepted: 09/29/2023] [Indexed: 10/19/2023] Open
Abstract
Distinct from the traditional diagnostic/prognostic biomarker (adopted as the indicator of disease state/process), the therapeutic biomarker (ThMAR) has emerged to be very crucial in the clinical development and clinical practice of all therapies. There are five types of ThMAR that have been found to play indispensable roles in various stages of drug discovery, such as: Pharmacodynamic Biomarker essential for guaranteeing the pharmacological effects of a therapy, Safety Biomarker critical for assessing the extent or likelihood of therapy-induced toxicity, Monitoring Biomarker indispensable for guiding clinical management by serially measuring patients' status, Predictive Biomarker crucial for maximizing the clinical outcome of a therapy for specific individuals, and Surrogate Endpoint fundamental for accelerating the approval of a therapy. However, these data of ThMARs has not been comprehensively described by any of the existing databases. Herein, a database, named 'TheMarker', was therefore constructed to (a) systematically offer all five types of ThMAR used at different stages of drug development, (b) comprehensively describe ThMAR information for the largest number of drugs among available databases, (c) extensively cover the widest disease classes by not just focusing on anticancer therapies. These data in TheMarker are expected to have great implication and significant impact on drug discovery and clinical practice, and it is freely accessible without any login requirement at: https://idrblab.org/themarker.
Collapse
Affiliation(s)
- Yintao Zhang
- College of Pharmaceutical Sciences, The First Affiliated Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Ying Zhou
- College of Pharmaceutical Sciences, The First Affiliated Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
- National Key Laboratory of Diagnosis and Treatment of Severe Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Yuan Zhou
- College of Pharmaceutical Sciences, The First Affiliated Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xinyuan Yu
- College of Pharmaceutical Sciences, The First Affiliated Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xinyi Shen
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven 06510, USA
| | - Yanfeng Hong
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Yuxin Zhang
- College of Pharmaceutical Sciences, The First Affiliated Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Shanshan Wang
- Qian Xuesen Collaborative Research Center of Astrochemistry and Space Life Sciences, Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
| | - Minjie Mou
- College of Pharmaceutical Sciences, The First Affiliated Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jinsong Zhang
- College of Pharmaceutical Sciences, The First Affiliated Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Lin Tao
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, The First Affiliated Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yunqing Qiu
- College of Pharmaceutical Sciences, The First Affiliated Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
- National Key Laboratory of Diagnosis and Treatment of Severe Infectious Disease, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Yuzong Chen
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, The Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen 518000, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, The First Affiliated Hospital, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| |
Collapse
|
8
|
Groves AM, Misra R, Clair G, Hernady E, Olson H, Orton D, Finkelstein J, Marples B, Johnston CJ. Influence of the irradiated pulmonary microenvironment on macrophage and T cell dynamics. Radiother Oncol 2023; 183:109543. [PMID: 36813173 PMCID: PMC10238652 DOI: 10.1016/j.radonc.2023.109543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/29/2022] [Accepted: 02/04/2023] [Indexed: 02/22/2023]
Abstract
BACKGROUND The lung is sensitive to radiation, increasing normal tissue toxicity risks following radiation therapy. Adverse outcomes include pneumonitis and pulmonary fibrosis, which result from dysregulated intercellular communication within the pulmonary microenvironment. Although macrophages are implicated in these pathogenic outcomes, the impact of their microenvironment is not well understood. MATERIALS AND METHODS C57BL/6J mice received 6Gyx5 irradiation to the right lung. Macrophage and T cell dynamics were investigated in ipsilateral right lungs, contralateral left lungs and non-irradiated control lungs 4-26wk post exposure. Lungs were evaluated by flow cytometry, histology and proteomics. RESULTS Following uni-lung irradiation, focal regions of macrophage accumulation were noted in both lungs by 8wk, however by 26wk fibrotic lesions were observed only in ipsilateral lungs. Infiltrating and alveolar macrophages populations expanded in both lungs, however transitional CD11b + alveolar macrophages persisted only in ipsilateral lungs and expressed lower CD206. Concurrently, arginase-1 + macrophages accumulated in ipsilateral but not contralateral lungs at 8 and 26wk post exposure, while CD206 + macrophages were absent from these accumulations. While radiation expanded CD8 + T cells in both lungs, T regulatory cells only increased in ipsilateral lungs. Unbiased proteomics analysis of immune cells revealed a substantial number of differentially expressed proteins in ipsilateral lungs when compared to contralateral lungs and both differed from non-irradiated controls. CONCLUSIONS Pulmonary macrophage and T cell dynamics are impacted by the microenvironmental conditions that develop following radiation exposure, both locally and systemically. While macrophages and T cells infiltrate and expand in both lungs, they diverge phenotypically depending on their environment.
Collapse
Affiliation(s)
- Angela M Groves
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.
| | - Ravi Misra
- Department of Pediatrics, Division of Neonatology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Geremy Clair
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Eric Hernady
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Heather Olson
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Danny Orton
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Jacob Finkelstein
- Department of Pediatrics, Division of Neonatology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Brian Marples
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Carl J Johnston
- Department of Pediatrics, Division of Neonatology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| |
Collapse
|
9
|
Biomarkers to Predict Lethal Radiation Injury to the Rat Lung. Int J Mol Sci 2023; 24:ijms24065627. [PMID: 36982722 PMCID: PMC10053311 DOI: 10.3390/ijms24065627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/25/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
Currently, there are no biomarkers to predict lethal lung injury by radiation. Since it is not ethical to irradiate humans, animal models must be used to identify biomarkers. Injury to the female WAG/RijCmcr rat has been well-characterized after exposure to eight doses of whole thorax irradiation: 0-, 5-, 10-, 11-, 12-, 13-, 14- and 15-Gy. End points such as SPECT imaging of the lung using molecular probes, measurement of circulating blood cells and specific miRNA have been shown to change after radiation. Our goal was to use these changes to predict lethal lung injury in the rat model, 2 weeks post-irradiation, before any symptoms manifest and after which a countermeasure can be given to enhance survival. SPECT imaging with 99mTc-MAA identified a decrease in perfusion in the lung after irradiation. A decrease in circulating white blood cells and an increase in five specific miRNAs in whole blood were also tested. Univariate analyses were then conducted on the combined dataset. The results indicated that a combination of percent change in lymphocytes and monocytes, as well as pulmonary perfusion volume could predict survival from radiation to the lungs with 88.5% accuracy (95% confidence intervals of 77.8, 95.3) with a p-value of < 0.0001 versus no information rate. This study is one of the first to report a set of minimally invasive endpoints to predict lethal radiation injury in female rats. Lung-specific injury can be visualized by 99mTc-MAA as early as 2 weeks after radiation.
Collapse
|
10
|
Shichijo K, Takatsuji T. Pathological observation of the effects of exposure to radioactive microparticles on experimental animals. JOURNAL OF RADIATION RESEARCH 2022; 63:i26-i37. [PMID: 35968993 PMCID: PMC9377041 DOI: 10.1093/jrr/rrac045] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/22/2022] [Indexed: 06/01/2023]
Abstract
Internal radiation exposure from neutron-induced radioisotopes that were environmentally activated following an atomic bombing or nuclear accident should be considered for a complete picture of the pathologic effects on survivors. Inhaled hot particles expose neighboring tissues to very high doses of particle beams, which can cause local tissue damage. Experimentally, a few μm of 55MnO2 powder was irradiated with neutrons at a nuclear reactor in order to generate 56MnO2 that emits β-rays. Rats were irradiated via inhalation. Pathological changes in various rat tissues were examined. In addition, the 56Mn β energy spectrum around the particles was calculated to determine the local dose rate and the cumulative dose. This review focuses on our latest pathological findings in lungs with internal radiation injury and discusses the pathological changes of early event damage caused by localized, very high-dose internal radiation exposure, including apoptosis, elastin stigma, emphysema, hemorrhage and severe inflammation. The pathological findings of lung tissue due to internal radiation exposure of 0.1 Gy were severe, with no pathological changes observed due to external exposure to γ radiation at a dose of 2.0 Gy. Therefore, it is suggested that new pathological analysis methods for internal exposure due to radioactive microparticles are required.
Collapse
Affiliation(s)
- Kazuko Shichijo
- Corresponding author. Department of Tumor and Diagnostic Pathology, Atomic Bomb Disease Institute, Nagasaki University, 112-4 Sakamoto, Nagasaki 852-8523, Japan. Tel.: +81-95-819-7107; Fax: +81-95-819-7108; E-mail:
| | - Toshihiro Takatsuji
- Faculty of Environmental Studies, Nagasaki University, Bunkyo 1-14, Nagasaki 852-8521, Japan
| |
Collapse
|
11
|
Bertho A, Dos Santos M, Braga-Cohen S, Buard V, Paget V, Guipaud O, Tarlet G, Milliat F, François A. Preclinical Model of Stereotactic Ablative Lung Irradiation Using Arc Delivery in the Mouse: Is Fractionation Worthwhile? Front Med (Lausanne) 2022; 8:794324. [PMID: 35004768 PMCID: PMC8739220 DOI: 10.3389/fmed.2021.794324] [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: 10/13/2021] [Accepted: 12/07/2021] [Indexed: 11/23/2022] Open
Abstract
Lung stereotactic body radiation therapy is characterized by a reduction in target volumes and the use of severely hypofractionated schedules. Preclinical modeling became possible thanks to rodent-dedicated irradiation devices allowing accurate beam collimation and focal lung exposure. Given that a great majority of publications use single dose exposures, the question we asked in this study was as follows: in incremented preclinical models, is it worth using fractionated protocols or should we continue focusing solely on volume limitation? The left lungs of C57BL/6JRj mice were exposed to ionizing radiation using arc therapy and 3 × 3 mm beam collimation. Three-fraction schedules delivered over a period of 1 week were used with 20, 28, 40, and 50 Gy doses per fraction. Lung tissue opacification, global histological damage and the numbers of type II pneumocytes and club cells were assessed 6 months post-exposure, together with the gene expression of several lung cells and inflammation markers. Only the administration of 3 × 40 Gy or 3 × 50 Gy generated focal lung fibrosis after 6 months, with tissue opacification visible by cone beam computed tomography, tissue scarring and consolidation, decreased club cell numbers and a reactive increase in the number of type II pneumocytes. A fractionation schedule using an arc-therapy-delivered three fractions/1 week regimen with 3 × 3 mm beam requires 40 Gy per fraction for lung fibrosis to develop within 6 months, a reasonable time lapse given the mouse lifespan. A comparison with previously published laboratory data suggests that, in this focal lung irradiation configuration, administering a Biological Effective Dose ≥ 1000 Gy should be recommended to obtain lung fibrosis within 6 months. The need for such a high dose per fraction challenges the appropriateness of using preclinical highly focused fractionation schedules in mice.
Collapse
Affiliation(s)
- Annaïg Bertho
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Morgane Dos Santos
- Laboratory of Radiobiology of Accidental Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Sarah Braga-Cohen
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Valérie Buard
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Vincent Paget
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Olivier Guipaud
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Georges Tarlet
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Fabien Milliat
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Agnès François
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| |
Collapse
|
12
|
Liu X, Shao C, Fu J. Promising Biomarkers of Radiation-Induced Lung Injury: A Review. Biomedicines 2021; 9:1181. [PMID: 34572367 PMCID: PMC8470495 DOI: 10.3390/biomedicines9091181] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/05/2021] [Accepted: 09/06/2021] [Indexed: 12/15/2022] Open
Abstract
Radiation-induced lung injury (RILI) is one of the main dose-limiting side effects in patients with thoracic cancer during radiotherapy. No reliable predictors or accurate risk models are currently available in clinical practice. Severe radiation pneumonitis (RP) or pulmonary fibrosis (PF) will reduce the quality of life, even when the anti-tumor treatment is effective for patients. Thus, precise prediction and early diagnosis of lung toxicity are critical to overcome this longstanding problem. This review summarizes the primary mechanisms and preclinical animal models of RILI reported in recent decades, and analyzes the most promising biomarkers for the early detection of lung complications. In general, ideal integrated models considering individual genetic susceptibility, clinical background parameters, and biological variations are encouraged to be built up, and more prospective investigations are still required to disclose the molecular mechanisms of RILI as well as to discover valuable intervention strategies.
Collapse
Affiliation(s)
- Xinglong Liu
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China;
| | - Chunlin Shao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China;
| | - Jiamei Fu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| |
Collapse
|
13
|
Yu H, Lam KO, Wu H, Green M, Wang W, Jin JY, Hu C, Jolly S, Wang Y, Kong FMS. Weighted-Support Vector Machine Learning Classifier of Circulating Cytokine Biomarkers to Predict Radiation-Induced Lung Fibrosis in Non-Small-Cell Lung Cancer Patients. Front Oncol 2021; 10:601979. [PMID: 33598430 PMCID: PMC7883680 DOI: 10.3389/fonc.2020.601979] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 12/08/2020] [Indexed: 01/06/2023] Open
Abstract
Background Radiation-induced lung fibrosis (RILF) is an important late toxicity in patients with non-small-cell lung cancer (NSCLC) after radiotherapy (RT). Clinically significant RILF can impact quality of life and/or cause non-cancer related death. This study aimed to determine whether pre-treatment plasma cytokine levels have a significant effect on the risk of RILF and investigate the abilities of machine learning algorithms for risk prediction. Methods This is a secondary analysis of prospective studies from two academic cancer centers. The primary endpoint was grade≥2 (RILF2), classified according to a system consistent with the consensus recommendation of an expert panel of the AAPM task for normal tissue toxicity. Eligible patients must have at least 6 months’ follow-up after radiotherapy commencement. Baseline levels of 30 cytokines, dosimetric, and clinical characteristics were analyzed. Support vector machine (SVM) algorithm was applied for model development. Data from one center was used for model training and development; and data of another center was applied as an independent external validation. Results There were 57 and 37 eligible patients in training and validation datasets, with 14 and 16.2% RILF2, respectively. Of the 30 plasma cytokines evaluated, SVM identified baseline circulating CCL4 as the most significant cytokine associated with RILF2 risk in both datasets (P = 0.003 and 0.07, for training and test sets, respectively). An SVM classifier predictive of RILF2 was generated in Cohort 1 with CCL4, mean lung dose (MLD) and chemotherapy as key model features. This classifier was validated in Cohort 2 with accuracy of 0.757 and area under the curve (AUC) of 0.855. Conclusions Using machine learning, this study constructed and validated a weighted-SVM classifier incorporating circulating CCL4 levels with significant dosimetric and clinical parameters which predicts RILF2 risk with a reasonable accuracy. Further study with larger sample size is needed to validate the role of CCL4, and this SVM classifier in RILF2.
Collapse
Affiliation(s)
- Hao Yu
- Biomedical Engineering, Shenzhen Polytechnic, Shenzhen, China.,BioHealth Informatics, School of Informatics and Computing, Indiana University - Purdue University Indianapolis (IUPUI), Indianapolis, IN, United States
| | - Ka-On Lam
- Department of Clinical Oncology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong.,Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Huanmei Wu
- BioHealth Informatics, School of Informatics and Computing, Indiana University - Purdue University Indianapolis (IUPUI), Indianapolis, IN, United States
| | - Michael Green
- Radiation Oncology, Ann Arbor VA Health Care, Ann Arbor, MI, United States.,Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
| | - Weili Wang
- University Hospitals, Cleveland Medical Center, Seidman Cancer Center and Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, United States
| | - Jian-Yue Jin
- University Hospitals, Cleveland Medical Center, Seidman Cancer Center and Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, United States
| | - Chen Hu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shruti Jolly
- Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
| | - Yang Wang
- Biomedical Engineering, Shenzhen Polytechnic, Shenzhen, China
| | - Feng-Ming Spring Kong
- Department of Clinical Oncology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong.,Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.,University Hospitals, Cleveland Medical Center, Seidman Cancer Center and Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
14
|
Impact of Local High Doses of Radiation by Neutron Activated Mn Dioxide Powder in Rat Lungs: Protracted Pathologic Damage Initiated by Internal Exposure. Biomedicines 2020; 8:biomedicines8060171. [PMID: 32586004 PMCID: PMC7345208 DOI: 10.3390/biomedicines8060171] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/17/2020] [Accepted: 06/21/2020] [Indexed: 11/24/2022] Open
Abstract
Internal radiation exposure from neutron-induced radioisotopes environmentally activated following atomic bombing or nuclear accidents should be considered for a complete picture of pathologic effects on survivors. Inhaled hot particles expose neighboring tissues to locally ultra-high doses of β-rays and can cause pathologic damage. 55MnO2 powder was activated by a nuclear reactor to make 56MnO2 which emits β-rays. Internal exposures were compared with external γ-rays. Male Wistar rats were administered activated powder by inhalation. Lung samples were observed by histological staining at six hours, three days, 14 days, two months, six months and eight months after the exposure. Synchrotron radiation—X-ray fluorescence—X-ray absorption near-edge structure (SR–XRF–XANES) was utilized for the chemical analysis of the activated 56Mn embedded in lung tissues. 56Mn beta energy spectrum around the particles was calculated to assess the local dose rate and accumulated dose. Hot particles located in the bronchiole and in damaged alveolar tissue were identified as accumulations of Mn and iron. Histological changes showed evidence of emphysema, hemorrhage and severe inflammation from six hours through eight months. Apoptosis was observed in the bronchiole epithelium. Our study shows early event damage from the locally ultra-high internal dose leads to pathogenesis. The trigger of emphysema and hemorrhage was likely early event damage to blood vessels integral to alveolar walls.
Collapse
|
15
|
Sunita, Sajid A, Singh Y, Shukla P. Computational tools for modern vaccine development. Hum Vaccin Immunother 2020; 16:723-735. [PMID: 31545127 PMCID: PMC7227725 DOI: 10.1080/21645515.2019.1670035] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/28/2019] [Accepted: 09/13/2019] [Indexed: 12/12/2022] Open
Abstract
Vaccines play an essential role in controlling the rates of fatality and morbidity. Vaccines not only arrest the beginning of different diseases but also assign a gateway for its elimination and reduce toxicity. This review gives an overview of the possible uses of computational tools for vaccine design. Moreover, we have described the initiatives of utilizing the diverse computational resources by exploring the immunological databases for developing epitope-based vaccines, peptide-based drugs, and other resources of immunotherapeutics. Finally, the applications of multi-graft and multivalent scaffolding, codon optimization and antibodyomics tools in identifying and designing in silico vaccine candidates are described.
Collapse
Affiliation(s)
- Sunita
- Enzyme Technology and Protein Bioinformatics Laboratory, Department of Microbiology, Maharshi Dayanand University, Rohtak, India
- Bacterial Pathogenesis Laboratory, Department of Zoology, University of Delhi, Delhi
| | - Andaleeb Sajid
- National Institutes of Health, National Cancer Institute, Bethesda, MD, USA
| | - Yogendra Singh
- Bacterial Pathogenesis Laboratory, Department of Zoology, University of Delhi, Delhi
| | - Pratyoosh Shukla
- Enzyme Technology and Protein Bioinformatics Laboratory, Department of Microbiology, Maharshi Dayanand University, Rohtak, India
| |
Collapse
|
16
|
Bao P, Zhao W, Mou M, Liu X. MicroRNA-21 mediates bone marrow mesenchymal stem cells protection of radiation-induced lung injury during the acute phase by regulating polarization of alveolar macrophages. Transl Cancer Res 2020; 9:231-239. [PMID: 35117177 PMCID: PMC8798259 DOI: 10.21037/tcr.2019.12.77] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/26/2019] [Indexed: 12/01/2022]
Abstract
Background Radiation-induced lung injury (RILI) often occurs in patients with non-small cell lung cancer (NSCLC) after radiotherapy, and the prognosis of patients with RILI is usually poor. This work plan to investigate the expression patterns of microRNA-21(miR-21) in NSCLC patients with RILI and the protective effects of miR-21 over-expressed bone marrow mesenchymal stem cells (BMSCs) against RILI in rat model. Methods MiR-21 expressions were determined in both serum samples and bronchoalveolar lavage fluid (BALF) samples from NSCLC patients after radiation therapy. The correlation between miR-21 expression and the follow-up clinical characterizations were determined. Further, miR-21 over-expressed BMSCs were transplanted into RILI rats and the protective effects were evaluated. BMSCs and alveolar macrophages (AMs) were co-cultured in vitro and the macrophage M1 polarization markers were determined by ELISA and qRT-PCR assays. Results Expression of miR-21 was significantly increased in NSCLC patients with RILI compared with control group, especially before or at 4 weeks after radiation therapy commenced. The miR-21 levels were highly correlated with IL-12, TNF-α, and IL-6 expressions and the severity of RILI. Animal based experiments demonstrated that BMSCs treatment had a remarkable effect on alleviating alveolitis in RILI rats, and miR-21 over-expression could enhance this effect significantly. Cell based experiments demonstrated that BMSCs notably inhibited M1 polarization of AMs and this inhibition is in a miR-21 dependent manner. Conclusions These results indicated that BMSCs could blocked the proinflammatory pathway of macrophage through miR-21 over-expression, thus could be a potential therapeutic strategy for RILI.
Collapse
Affiliation(s)
- Pengtao Bao
- Department of Respiration, the Eighth Medical Center of PLA General Hospital, Beijing 100091, China
| | - Weiguo Zhao
- Department of Respiration, the Eighth Medical Center of PLA General Hospital, Beijing 100091, China
| | - Mi Mou
- Department of Respiration, the Eighth Medical Center of PLA General Hospital, Beijing 100091, China
| | - Xiaofei Liu
- Department of Respiration, the Eighth Medical Center of PLA General Hospital, Beijing 100091, China
| |
Collapse
|
17
|
Stirling ER, Cook KL, Roberts DD, Soto-Pantoja DR. Metabolomic Analysis Reveals Unique Biochemical Signatures Associated with Protection from Radiation Induced Lung Injury by Lack of cd47 Receptor Gene Expression. Metabolites 2019; 9:E218. [PMID: 31597291 PMCID: PMC6835245 DOI: 10.3390/metabo9100218] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/20/2019] [Accepted: 09/30/2019] [Indexed: 01/10/2023] Open
Abstract
The goal of this study was to interrogate biochemical profiles manifested in mouse lung tissue originating from wild type (WT) and cd47 null mice with the aim of revealing the in vivo role of CD47 in the metabolic response to ionizing radiation, especially changes related to the known association of CD47 deficiency with increased tissue viability and survival. For this objective, we performed global metabolomic analysis in mouse lung tissue collected from (C57Bl/6 background) WT and cd47 null mice with and without exposure to 7.6 Gy whole body radiation. Principal component analysis and hierarchical clustering revealed a consistent separation between genotypes following radiation exposure. Random forest analysis also revealed a unique biochemical signature in WT and cd47 null mice following treatment. Our data show that cd47 null irradiated lung tissue activates a unique set of metabolic pathways that facilitate the handling of reactive oxygen species, lipid metabolism, nucleotide metabolism and nutrient metabolites which may be regulated by microbial processing. Given that cd47 has pleiotropic effects on responses to ionizing radiation, we not only propose this receptor as a therapeutic target but postulate that the biomarkers regulated in this study associated with radioprotection are potential mitigators of radiation-associated pathologies, including the onset of pulmonary disease.
Collapse
Affiliation(s)
- Elizabeth R Stirling
- Department of Cancer Biology, Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27101, USA.
- Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27101, USA.
| | - Katherine L Cook
- Department of Cancer Biology, Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27101, USA.
- Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27101, USA.
- Department of Surgery, Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27101, USA.
| | - David D Roberts
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - David R Soto-Pantoja
- Department of Cancer Biology, Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27101, USA.
- Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27101, USA.
- Department of Surgery, Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27101, USA.
- Department of Radiation Oncology, Wake Forest School of Medicine Comprehensive Cancer Center, Winston-Salem, NC 27101, USA.
| |
Collapse
|
18
|
Hanson KM, Hernady EB, Reed CK, Johnston CJ, Groves AM, Finkelstein JN. Apoptosis Resistance in Fibroblasts Precedes Progressive Scarring in Pulmonary Fibrosis and Is Partially Mediated by Toll-Like Receptor 4 Activation. Toxicol Sci 2019; 170:489-498. [PMID: 31020321 PMCID: PMC6657580 DOI: 10.1093/toxsci/kfz103] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Inhalation of environmental toxicants such as cigarette smoke, metal or wood dust, silica, or asbestos is associated with increased risk for idiopathic pulmonary fibrosis (IPF). IPF involves progressive scarring of lung tissue, which interferes with normal respiration and is ultimately fatal; however, the complex cellular mechanisms of IPF pathogenesis remain unclear. Fibroblast apoptosis is essential in normal wound healing but is dysregulated in IPF. Recent studies suggest that Toll-like receptor 4 (TLR4) is key in the onset of IPF. Here, radiation-induced PF was used as a model for IPF because it very closely mimics the progressive and intractable nature of IPF. Female C57BL/6J (C57) and C57BL/6J TLR4-/- mice were exposed to a single dose of 13 Gy whole-thorax ionizing radiation. Although both strains showed similar levels of immediate radiation-induced damage, C57 mice exhibited more extensive fibrosis at 22-week postirradiation (PI) than TLR4-/- mice. Isolated C57 primary 1° MLFs showed decreased apoptosis susceptibility as early as 8-week postirradiation, a phenotype that persisted for the remainder of the radiation response. TLR4-/- 1° mouse lung fibroblasts did not exhibit significant apoptosis resistance at any point. Systemic release of high mobility group box 1, a TLR4 agonist, during the pneumonitis phase of the radiation response may act through TLR4 to contribute to fibroblast apoptosis resistance and thus interfere with wound resolution. These findings demonstrate that apoptosis resistance occurs earlier in pulmonary fibrosis pathogenesis than previously assumed, and that TLR4 signaling is a key mediator in this process.
Collapse
Affiliation(s)
| | | | - Christina K Reed
- Department of Pediatrics and Neonatology, University of Rochester Medical Center, Rochester, New York 14642
| | - Carl J Johnston
- Department of Pediatrics and Neonatology, University of Rochester Medical Center, Rochester, New York 14642
| | - Angela M Groves
- Department of Pediatrics and Neonatology, University of Rochester Medical Center, Rochester, New York 14642
| | - Jacob N Finkelstein
- Department of Environmental Medicine
- Department of Radiation Oncology
- Department of Pediatrics and Neonatology, University of Rochester Medical Center, Rochester, New York 14642
| |
Collapse
|
19
|
MacVittie TJ, Farese AM, Kane MA. ARS, DEARE, and Multiple-organ Injury: A Strategic and Tactical Approach to Link Radiation Effects, Animal Models, Medical Countermeasures, and Biomarker Development to Predict Clinical Outcome. HEALTH PHYSICS 2019; 116:297-304. [PMID: 30608246 PMCID: PMC8439279 DOI: 10.1097/hp.0000000000001045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Affiliation(s)
| | - Ann M Farese
- University of Maryland School of Medicine, Baltimore, MD
| | - Maureen A Kane
- University of Maryland School of Pharmacy, Baltimore, MD
| |
Collapse
|