1
|
Bourgois A, Cosler G, Riccobono D, Le Gallic C, François S, Van der Meeren A. DTPA and anti-inflammatory drug associations to alleviate Pu-induced response of macrophages in vitro. Toxicol In Vitro 2025; 104:106007. [PMID: 39837393 DOI: 10.1016/j.tiv.2025.106007] [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: 09/20/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 01/23/2025]
Abstract
Internal contamination by inhalation of plutonium poorly soluble compounds leads to their long time retention in alveolar macrophages inducing delayed pathology development. As previous studies highlighted co-localization of retained Pu and inflammatory lesions, this study was designed to assess the combined effect of the reference treatment (DTPA) and anti-inflammatory drugs on Pu-induced early response of macrophages in vitro. Pu colloids, mimicking poorly soluble Pu, were characterized using filtration and solid-state nuclear track detectors CR39. Their bioavailability was determined using a biphasic acellular model. Treatment effects on Pu dissolution and release as well as on Pu-induced pro-inflammatory response were assessed over 7 days on macrophage-like cells. DTPA treatment, associated or not with anti-inflammatory drug, increased Pu dissolution and release from contaminated THP-1 differentiated cells after 7 days. Significant decreases in Pu-induced IL-8 and MCP-1 secretions were also observed with anti-inflammatory treatment associated or not with DTPA. This study highlighted the ability of DTPA to partially dissolve a poorly soluble form of Pu as well as the ability of anti-inflammatory drugs to modulate Pu-induced pro-inflammatory response in macrophage-like cells. These treatments seem a promising strategy to improve the clinical management of Pu pulmonary contaminations and to limit delayed pulmonary pathology occurrence.
Collapse
Affiliation(s)
- Alexandra Bourgois
- French Armed Forces Biomedical Research Institute (IRBA), Radiation Biological Effects Department, Brétigny-sur-Orge, France.
| | - Guillaume Cosler
- French Armed Forces Biomedical Research Institute (IRBA), Radiation Biological Effects Department, Brétigny-sur-Orge, France
| | - Diane Riccobono
- French Armed Forces Biomedical Research Institute (IRBA), Radiation Biological Effects Department, Brétigny-sur-Orge, France; INSERM 1296: Radiation, Defense, Health and Environment, Lyon and Brétigny-sur-Orge, France
| | - Clélia Le Gallic
- French Armed Forces Biomedical Research Institute (IRBA), Radiation Biological Effects Department, Brétigny-sur-Orge, France; Present address: French Government Defense Procurement and Technology Agency (DGA), CBRN Control, Vert-le-Petit, France
| | - Sabine François
- French Armed Forces Biomedical Research Institute (IRBA), Radiation Biological Effects Department, Brétigny-sur-Orge, France; INSERM 1296: Radiation, Defense, Health and Environment, Lyon and Brétigny-sur-Orge, France
| | - Anne Van der Meeren
- Atomic Energy and Alternative Energies Commission (CEA), Laboratory of Radiotoxicology, CEA, Paris-Saclay University, Bruyères-le-Châtel, France
| |
Collapse
|
2
|
Suri C, Pande B, Sahithi LS, Sahu T, Verma HK. Interplay between Lung Diseases and Viral Infections: A Comprehensive Review. Microorganisms 2024; 12:2030. [PMID: 39458339 PMCID: PMC11510474 DOI: 10.3390/microorganisms12102030] [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: 08/25/2024] [Revised: 09/16/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
The intricate relationship between chronic lung diseases and viral infections is a significant concern in respiratory medicine. We explore how pre-existing lung conditions, including chronic obstructive pulmonary disease, asthma, and interstitial lung diseases, influence susceptibility, severity, and outcomes of viral infections. We also examine how viral infections exacerbate and accelerate the progression of lung disease by disrupting immune responses and triggering inflammatory pathways. By summarizing current evidence, this review highlights the bidirectional nature of these interactions, where underlying lung diseasesincrease vulnerability to viral infections, while these infections, in turn, worsen the clinical course. This review underscores the importance of preventive measures, such as vaccination, early detection, and targeted therapies, to mitigate adverse outcomes in patients with chronic lung conditions. The insights provided aim to inform clinical strategies that can improve patient management and reduce the burden of chronic lung diseases exacerbated by viral infections.
Collapse
Affiliation(s)
- Chahat Suri
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada;
| | - Babita Pande
- Department of Physiology, All India Institute of Medical Sciences, Raipur 492099, India; (B.P.); (T.S.)
| | | | - Tarun Sahu
- Department of Physiology, All India Institute of Medical Sciences, Raipur 492099, India; (B.P.); (T.S.)
| | - Henu Kumar Verma
- Department of Immunopathology, Institute of Lungs Health and Immunity, Comprehensive Pneumology Center, Helmholtz Zentrum, Neuherberg, 85764 Munich, Germany
| |
Collapse
|
3
|
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
|
4
|
Mohammed AI, Fedoruk L, Fisher N, Liu AX, Khanna S, Naylor K, Gong Z, Celentano A, Alrashdan MS, Cirillo N. Systemic Anti-Inflammatory Agents in the Prevention of Chemoradiation-Induced Mucositis: A Review of Randomised Controlled Trials. Biomolecules 2024; 14:560. [PMID: 38785967 PMCID: PMC11117894 DOI: 10.3390/biom14050560] [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: 03/13/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024] Open
Abstract
Mucositis is a pathological condition characterised by inflammation and ulceration of the mucous membranes lining the alimentary canal, particularly in the mouth (oral mucositis) and the gastrointestinal tract. It is a common side effect of cancer treatments, including chemotherapy and radiotherapy, and it is sometimes responsible for treatment interruptions. Preventing mucositis throughout the alimentary tract is therefore crucial. However, current interventions mainly target either oral or gastrointestinal side effects. This review aimed to investigate the use of systemically administered anti-inflammatory agents to prevent mucositis in cancer patients undergoing cancer treatment. PubMed, Ovid, Scopus, Web of Science, WHO ICTRP and ClinicalTrials.gov were screened to identify eligible randomised controlled trials (RCTs). The published literature on anti-inflammatory agents provides mixed evidence regarding the degree of efficacy in preventing/reducing the severity of mucositis in most anticancer treatments; however, sample size continued to be a significant limitation, alongside others discussed. Our review yielded a list of several anti-inflammatory agents that exhibit potential mucositis-preventive effects in cancer patients undergoing cancer treatment, which can be used to inform clinical practice.
Collapse
Affiliation(s)
- Ali I. Mohammed
- Melbourne Dental School, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Carlton, VIC 3053, Australia; (A.I.M.); (L.F.); (N.F.); (A.X.L.); (S.K.); (K.N.); (Z.G.); (A.C.)
| | - Lexi Fedoruk
- Melbourne Dental School, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Carlton, VIC 3053, Australia; (A.I.M.); (L.F.); (N.F.); (A.X.L.); (S.K.); (K.N.); (Z.G.); (A.C.)
| | - Nicholas Fisher
- Melbourne Dental School, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Carlton, VIC 3053, Australia; (A.I.M.); (L.F.); (N.F.); (A.X.L.); (S.K.); (K.N.); (Z.G.); (A.C.)
| | - Andy Xiaoqian Liu
- Melbourne Dental School, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Carlton, VIC 3053, Australia; (A.I.M.); (L.F.); (N.F.); (A.X.L.); (S.K.); (K.N.); (Z.G.); (A.C.)
| | - Samar Khanna
- Melbourne Dental School, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Carlton, VIC 3053, Australia; (A.I.M.); (L.F.); (N.F.); (A.X.L.); (S.K.); (K.N.); (Z.G.); (A.C.)
| | - Kaelan Naylor
- Melbourne Dental School, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Carlton, VIC 3053, Australia; (A.I.M.); (L.F.); (N.F.); (A.X.L.); (S.K.); (K.N.); (Z.G.); (A.C.)
| | - Ziyi Gong
- Melbourne Dental School, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Carlton, VIC 3053, Australia; (A.I.M.); (L.F.); (N.F.); (A.X.L.); (S.K.); (K.N.); (Z.G.); (A.C.)
| | - Antonio Celentano
- Melbourne Dental School, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Carlton, VIC 3053, Australia; (A.I.M.); (L.F.); (N.F.); (A.X.L.); (S.K.); (K.N.); (Z.G.); (A.C.)
| | - Mohammad S. Alrashdan
- Department of Oral and Craniofacial Health Sciences, College of Dental Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates;
- Department of Oral Medicine and Oral Surgery, Faculty of Dentistry, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Nicola Cirillo
- Melbourne Dental School, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Carlton, VIC 3053, Australia; (A.I.M.); (L.F.); (N.F.); (A.X.L.); (S.K.); (K.N.); (Z.G.); (A.C.)
- School of Dentistry, University of Jordan, Amman 11942, Jordan
| |
Collapse
|
5
|
Abston E, Zhou IY, Saenger JA, Shuvaev S, Akam E, Esfahani SA, Hariri LP, Rotile NJ, Crowley E, Montesi SB, Humblet V, Arabasz G, Khandekar M, Catana C, Fintelmann FJ, Caravan P, Lanuti M. Noninvasive Quantification of Radiation-Induced Lung Injury Using a Targeted Molecular Imaging Probe. Int J Radiat Oncol Biol Phys 2024; 118:1228-1239. [PMID: 38072325 PMCID: PMC11184492 DOI: 10.1016/j.ijrobp.2023.11.032] [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: 06/20/2023] [Revised: 11/13/2023] [Accepted: 11/17/2023] [Indexed: 12/19/2023]
Abstract
PURPOSE Radiation-induced lung injury (RILI) is a progressive inflammatory process seen after irradiation for lung cancer. The disease can be insidious, often characterized by acute pneumonitis followed by chronic fibrosis with significant associated morbidity. No therapies are approved for RILI, and accurate disease quantification is a major barrier to improved management. Here, we sought to noninvasively quantify RILI using a molecular imaging probe that specifically targets type 1 collagen in mouse models and patients with confirmed RILI. METHODS AND MATERIALS Using a murine model of lung radiation, mice were imaged with EP-3533, a type 1 collagen probe, to characterize the development of RILI and to assess disease mitigation after losartan treatment. The human analog probe 68Ga-CBP8, targeting type 1 collagen, was tested on excised human lung tissue containing RILI and was quantified via autoradiography. 68Ga-CBP8 positron emission tomography was used to assess RILI in vivo in 6 human subjects. RESULTS Murine models demonstrated that probe signal correlated with progressive RILI severity over 6 months. The probe was sensitive to mitigation of RILI by losartan. Excised human lung tissue with RILI had increased binding versus unirradiated control tissue, and 68Ga-CBP8 uptake correlated with collagen proportional area. Human imaging revealed significant 68Ga-CBP8 uptake in areas of RILI and minimal background uptake. CONCLUSIONS These findings support the ability of a molecular imaging probe targeted at type 1 collagen to detect RILI in preclinical models and human disease, suggesting a role for targeted molecular imaging of collagen in the assessment of RILI.
Collapse
Affiliation(s)
- Eric Abston
- Division of Thoracic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Iris Y Zhou
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts; Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, Massachusetts
| | - Jonathan A Saenger
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sergey Shuvaev
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts; Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, Massachusetts
| | - Eman Akam
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts
| | - Shadi A Esfahani
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Lida P Hariri
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nicholas J Rotile
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts; Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, Massachusetts
| | - Elizabeth Crowley
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sydney B Montesi
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Grae Arabasz
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts
| | - Melin Khandekar
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ciprian Catana
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts; Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, Massachusetts
| | - Florian J Fintelmann
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts; Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Institute for Innovation in Imaging, Massachusetts General Hospital, Boston, Massachusetts
| | - Michael Lanuti
- Division of Thoracic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
6
|
Zhu KY, Karimi AH, Lavu M, Burkhart RJ, Kamath AF. Impact of external beam radiation on total shoulder arthroplasty outcomes: a propensity-matched cohort study. Arch Orthop Trauma Surg 2024; 144:113-119. [PMID: 37670152 DOI: 10.1007/s00402-023-05048-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/26/2023] [Indexed: 09/07/2023]
Abstract
BACKGROUND External beam radiation therapy has a number of deleterious effects on the body, and a number of post-operative complications have been reported for several surgeries including total knee arthroplasty. However, few studies have investigated the impact of external beam radiation therapy for total shoulder arthroplasty (TSA). Our study aimed to assess the systemic and joint complications associated with TSA in patients with prior radiation exposures, as well as evaluate the surgical outcomes of radiation patients compared to non-radiation TSA patients. MATERIALS AND METHODS A retrospective cohort analysis was conducted using the TriNetX Analytics Network. A 1:1 propensity score matching function was utilized to create two cohorts with matched baseline characteristics within the TriNetX network. Comparisons of the primary and secondary outcomes between the two cohorts were made using odds ratios. A p value of < 0.05 was determined to be significant. RESULTS A total of 75,510 patients that received TSA were identified with 1505 having a history of radiation therapy (RT) and 73,605 with no radiation therapy (non-RT). After propensity matching, both groups contained 1484 patients. RT patients were at higher risk for developing prosthetic joint infection, acute renal failure, altered mental state, cerebrovascular event, DVT, PE, pneumonia, respiratory failure, and UTI compared to non-RT patients at different time points (p < 0.5). CONCLUSION Patients with prior history of external beam radiation undergoing TSA had a higher risk of systemic complications and prosthetic joint infection compared to patients without a prior history. These complications suggest a more complicated post-operative management course for these patients.
Collapse
Affiliation(s)
- Kevin Y Zhu
- Department of Orthopedic Surgery, Center for Hip Preservation, Cleveland Clinic Foundation, 9500 Euclid Ave, Mail Code A41, Cleveland, OH, 44195, USA
| | - Amir H Karimi
- Department of Orthopedic Surgery, Center for Hip Preservation, Cleveland Clinic Foundation, 9500 Euclid Ave, Mail Code A41, Cleveland, OH, 44195, USA
| | - Monish Lavu
- Department of Orthopedic Surgery, Center for Hip Preservation, Cleveland Clinic Foundation, 9500 Euclid Ave, Mail Code A41, Cleveland, OH, 44195, USA
| | - Robert J Burkhart
- Department of Orthopedic Surgery, Center for Hip Preservation, Cleveland Clinic Foundation, 9500 Euclid Ave, Mail Code A41, Cleveland, OH, 44195, USA
| | - Atul F Kamath
- Department of Orthopedic Surgery, Center for Hip Preservation, Cleveland Clinic Foundation, 9500 Euclid Ave, Mail Code A41, Cleveland, OH, 44195, USA.
| |
Collapse
|
7
|
Sakane T, Nakajima K, Iwata H, Nakano T, Hagui E, Oguri M, Nomura K, Hattori Y, Ogino H, Haneda H. Lobectomy versus proton therapy for stage I non-small cell lung cancer. J Thorac Cardiovasc Surg 2023; 166:1490-1501.e2. [PMID: 37625619 DOI: 10.1016/j.jtcvs.2023.08.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/02/2023] [Accepted: 08/13/2023] [Indexed: 08/27/2023]
Abstract
OBJECTIVE Lobectomy is the standard treatment for patients with early-stage non-small cell lung cancer (NSCLC). In recent years, an increasing number of patients with lung cancer have been treated using proton therapy (PT). We conducted a propensity score-matched analysis to compare the treatment outcomes of these 2 modalities. METHODS We retrospectively reviewed data from 275 patients with histologically confirmed clinical stage I NSCLC who underwent lobectomy (n = 206) or PT (n = 69) at our institution from July 2013 to December 2020. The end points were overall survival (OS), cause-specific survival, recurrence-free survival (RFS), local control, regional lymph node control, and distant control. Propensity score matching was performed to reduce selection bias in the 2 groups. RESULTS The matched cohort consisted of 59 patients who underwent lobectomy and 59 patients who underwent PT with a median follow-up period of 50 months. There were no significant differences in OS (P = .26), cause-specific survival (P = .33), RFS (P = .53), local control (P = .41), regional lymph node control (P = .98), and distant control (P = .31). In the lobectomy and PT groups, the 5-year OS rate was 85.8% and 79.1%, respectively, the RFS rate was 82.3% and 77.8%, and the local control rate was 92.1% and 96.6%. CONCLUSIONS We found no difference in survival or disease control between lobectomy and PT in patients with histologically confirmed clinical stage I NSCLC. Despite these findings, the potential for unmeasured confounding factors remains, and randomized control trials are needed to better compare these treatment modalities.
Collapse
Affiliation(s)
- Tadashi Sakane
- Department of Thoracic Surgery, Nagoya City University West Medical Center, Nagoya, Japan.
| | - Koichiro Nakajima
- Department of Radiation Oncology, Nagoya Proton Therapy Center, Nagoya City University West Medical Center, Nagoya, Japan
| | - Hiromitsu Iwata
- Department of Radiation Oncology, Nagoya Proton Therapy Center, Nagoya City University West Medical Center, Nagoya, Japan
| | - Tomoharu Nakano
- Department of Thoracic Surgery, Nagoya City University West Medical Center, Nagoya, Japan
| | - Emi Hagui
- Department of Thoracic Surgery, Nagoya City University West Medical Center, Nagoya, Japan
| | - Masanosuke Oguri
- Department of Radiation Oncology, Nagoya Proton Therapy Center, Nagoya City University West Medical Center, Nagoya, Japan
| | - Kento Nomura
- Department of Radiation Oncology, Nagoya Proton Therapy Center, Nagoya City University West Medical Center, Nagoya, Japan
| | - Yukiko Hattori
- Department of Radiation Oncology, Nagoya Proton Therapy Center, Nagoya City University West Medical Center, Nagoya, Japan
| | - Hiroyuki Ogino
- Department of Radiation Oncology, Nagoya Proton Therapy Center, Nagoya City University West Medical Center, Nagoya, Japan
| | - Hiroshi Haneda
- Department of Thoracic Surgery, Nagoya City University West Medical Center, Nagoya, Japan
| |
Collapse
|
8
|
Hurley K, Clow R, Jadhav A, Azzam EI, Wang Y. Mitigation of acute radiation syndrome (ARS) with human umbilical cord blood. Int J Radiat Biol 2023; 100:317-334. [PMID: 37967239 DOI: 10.1080/09553002.2023.2277372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/27/2023] [Indexed: 11/17/2023]
Abstract
PURPOSE The growing concern over potential unintended nuclear accidents or malicious activities involving nuclear/radiological devices cannot be overstated. Exposure to whole-body doses of radiation can result in acute radiation syndrome (ARS), colloquially known as "radiation sickness," which can severely damage various organ systems. Long-term health consequences, such as cancer and cardiovascular disease, can develop many years post-exposure. Identifying effective medical countermeasures and devising a strategic medical plan represents an urgent, unmet need. Various clinical studies have investigated the therapeutic use of umbilical cord blood (UCB) for a range of illnesses, including ARS. The objective of this review is to thoroughly discuss ARS and its sub-syndromes, and to highlight recent findings regarding the use of UCB for radiation injury. UCB, a rich source of stem cells, boasts numerous advantages over other stem cell sources, like bone marrow, owing to its ease of collection and relatively low risk of severe graft-versus-host disease. Preclinical studies suggest that treatment with UCB, and often UCB-derived mesenchymal stromal cells (MSCs), results in improved survival, accelerated hematopoietic recovery, reduced gastrointestinal tract damage, and mitigation of radiation-induced pneumonitis and pulmonary fibrosis. Interestingly, recent evidence suggests that UCB-derived exosomes and their microRNAs (miRNAs) might assist in treating radiation-induced damage, largely by inhibiting fibrotic pathways. CONCLUSION UCB holds substantial potential as a radiation countermeasure, and future research should focus on establishing treatment parameters for ARS victims.
Collapse
Affiliation(s)
- Kate Hurley
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, Canada
| | - Rachel Clow
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, Canada
| | - Ashok Jadhav
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, Canada
| | - Edouard I Azzam
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, Canada
| | - Yi Wang
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
| |
Collapse
|
9
|
Hyun D, Han SJ, Ji W, Choi C, Lee JC, Kim HC. Clinical characteristics and prognostic impact of acute exacerbations in patients with interstitial lung disease and lung cancer: A single-center, retrospective cohort study. Thorac Cancer 2023; 14:3323-3330. [PMID: 37772425 PMCID: PMC10665778 DOI: 10.1111/1759-7714.15124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/17/2023] [Accepted: 09/19/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Although acute exacerbation (AE) after treatment for lung cancer (LC) is a poor prognostic factor in patients with interstitial lung disease associated with lung cancer (ILD-LC), the risk of AE according to cancer treatment type remains unclear. Therefore, in the present study, we aimed to investigate the association between AE and treatment received for LC in patients with ILD-LC. METHODS We conducted a retrospective study of patients with ILD-LC who had undergone treatment for LC between January 2018 and December 2022. The primary study outcome was the incidence of AE within 12 months of treatment for LC according to treatment type. The association between AE and all-cause mortality was evaluated as a secondary outcome. RESULTS Among a total of 137 patients, 23 (16.8%) developed AE within 12 months of treatment for LC. The incidence of AE according to treatment type was 4.3% for surgery, 16.2% for radiotherapy, 15.6% for chemotherapy, and 54.5% for concurrent chemoradiation therapy (CCRT). Patients who received CCRT were more likely to develop AE, even after adjustment for covariables (hazard ratio [HR], 15.39; 95% confidence interval [CI]: 4.00-59.19; p < 0.001). In addition, AE within 12 months of treatment for LC was associated with an increased risk of all-cause mortality (HR, 2.82; 95% CI: 1.13-7.04; p = 0.026). CONCLUSION Among treatment options for patients with ILD-LC, CCRT was associated with an increased risk for AE. In addition, patients with AE had a higher mortality rate than patients without AE.
Collapse
Affiliation(s)
- Dong‐gon Hyun
- Department of Pulmonary and Critical Care Medicine, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| | - Soo Jin Han
- Department of Pulmonary and Critical Care Medicine, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| | - Wonjun Ji
- Department of Pulmonary and Critical Care Medicine, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| | - Chang‐Min Choi
- Department of Pulmonary and Critical Care Medicine, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
- Department of Oncology, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| | - Jae Cheol Lee
- Department of Oncology, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| | - Ho Cheol Kim
- Department of Pulmonary and Critical Care Medicine, Asan Medical CenterUniversity of Ulsan College of MedicineSeoulRepublic of Korea
| |
Collapse
|
10
|
Cassatt DR, Winters TA, PrabhuDas M. Immune Dysfunction from Radiation Exposure. Radiat Res 2023; 200:389-395. [PMID: 37702416 PMCID: PMC10599297 DOI: 10.1667/rade-22-00197.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 08/14/2023] [Indexed: 09/14/2023]
Abstract
Exposure to ionizing radiation causes acute damage and loss of bone marrow and peripheral immune cells that can result in high mortality due to reduced resistance to infections and hemorrhage. Besides these acute effects, tissue damage from radiation can trigger inflammatory responses, leading to progressive and chronic tissue damage by radiation-induced loss of immune cell types that are required for resolving tissue injuries. Understanding the mechanisms involved in radiation-induced immune system injury and repair will provide new insights for developing medical countermeasures that help restore immune homeostasis. For these reasons, The Radiation and Nuclear Countermeasures Program (RNCP) and the Basic Immunology Branch (BIB) under the Division of Allergy, Immunology, and Transplantation (DAIT) within the National Institute of Allergy and Infectious Diseases (NIAID) convened a two-day workshop, along with partners from the Biomedical Advanced Research and Development Authority (BARDA), and the Radiation Injury Treatment Network (RITN). This workshop, titled "Immune Dysfunction from Radiation Exposure," was held virtually on September 9-10, 2020; this Commentary provides a high-level overview of what was discussed at the meeting.
Collapse
Affiliation(s)
- David R. Cassatt
- Radiation and Nuclear Countermeasures Program (RNCP), Rockville, Maryland
| | - Thomas A. Winters
- Radiation and Nuclear Countermeasures Program (RNCP), Rockville, Maryland
| | - Mercy PrabhuDas
- Basic Immunology Branch (BIB), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| |
Collapse
|
11
|
Hollingsworth BA, Aldrich JT, Case CM, DiCarlo AL, Hoffman CM, Jakubowski AA, Liu Q, Loelius SG, PrabhuDas M, Winters TA, Cassatt DR. Immune Dysfunction from Radiation Exposure. Radiat Res 2023; 200:396-416. [PMID: 38152282 PMCID: PMC10751071 DOI: 10.1667/rade-22-00004.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
The hematopoietic system is highly sensitive to ionizing radiation. Damage to the immune system may result in opportunistic infections and hemorrhage, which could lead to mortality. Inflammation triggered by tissue damage can also lead to additional local or widespread tissue damage. The immune system is responsible for tissue repair and restoration, which is made more challenging when it is in the process of self-recovery. Because of these challenges, the Radiation and Nuclear Countermeasures Program (RNCP) and the Basic Immunology Branch (BIB) under the Division of Allergy, Immunology, and Transplantation (DAIT) within the National Institute of Allergy and Infectious Diseases (NIAID), along with partners from the Biomedical Advanced Research and Development Authority (BARDA), and the Radiation Injury Treatment Network (RITN) sponsored a two-day meeting titled Immune Dysfunction from Radiation Exposure held on September 9-10, 2020. The intent was to discuss the manifestations and mechanisms of radiation-induced immune dysfunction in people and animals, identify knowledge gaps, and discuss possible treatments to restore immune function and enhance tissue repair after irradiation.
Collapse
Affiliation(s)
- Brynn A. Hollingsworth
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
- Current address: Center for Biologics Evaluation and Research (CBER), Food and Drug Administration (FDA), Silver Spring, Maryland
| | | | - Cullen M. Case
- Radiation Injury Treatment Network, Minneapolis, Minnesota
| | - Andrea L. DiCarlo
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Corey M. Hoffman
- Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS), Washington, DC
| | | | - Qian Liu
- Basic Immunology Branch (BIB), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Shannon G. Loelius
- Biomedical Advanced Research and Development Authority (BARDA), Office of the Assistant Secretary for Preparedness and Response (ASPR), Department of Health and Human Services (HHS), Washington, DC
| | - Mercy PrabhuDas
- Basic Immunology Branch (BIB), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - Thomas A. Winters
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| | - David R. Cassatt
- Radiation and Nuclear Countermeasures Program (RNCP), Division of Allergy, Immunology and Transplantation (DAIT), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland
| |
Collapse
|
12
|
Abston E, Zhou IY, Saenger JA, Shuvaev S, Akam E, Esfahani SA, Hariri LP, Rotile NJ, Crowley E, Montesi SB, Humblet V, Arabasz G, Catana C, Fintelmann FJ, Caravan P, Lanuti M. Noninvasive Quantification of Radiation-Induced Lung Injury using a Targeted Molecular Imaging Probe. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.25.23295897. [PMID: 37808864 PMCID: PMC10557816 DOI: 10.1101/2023.09.25.23295897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Rationale Radiation-induced lung injury (RILI) is a progressive inflammatory process commonly seen following irradiation for lung cancer. The disease can be insidious, often characterized by acute pneumonitis followed by chronic fibrosis with significant associated morbidity. No therapies are approved for RILI, and accurate disease quantification is a major barrier to improved management. Objective To noninvasively quantify RILI, utilizing a molecular imaging probe that specifically targets type 1 collagen in mouse models and patients with confirmed RILI. Methods Using a murine model of lung radiation, mice were imaged with EP-3533, a type 1 collagen probe to characterize the development of RILI and to assess disease mitigation following losartan treatment. The human analog probe targeted against type 1 collagen, 68Ga-CBP8, was tested on excised human lung tissue containing RILI and quantified via autoradiography. Finally, 68Ga-CBP8 PET was used to assess RILI in vivo in six human subjects. Results Murine models demonstrated that probe signal correlated with progressive RILI severity over six-months. The probe was sensitive to mitigation of RILI by losartan. Excised human lung tissue with RILI had increased binding vs unirradiated control tissue and 68Ga-CBP8 uptake correlated with collagen proportional area. Human imaging revealed significant 68Ga-CBP8 uptake in areas of RILI and minimal background uptake. Conclusions These findings support the ability of a molecular imaging probe targeted at type 1 collagen to detect RILI in preclinical models and human disease, suggesting a role for targeted molecular imaging of collagen in the assessment of RILI.Clinical trial registered with www.clinicaltrials.gov (NCT04485286, NCT03535545).
Collapse
Affiliation(s)
- Eric Abston
- Division of Thoracic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Iris Y Zhou
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
- The Institute for Innovation in Imaging Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jonathan A Saenger
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Sergey Shuvaev
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
- The Institute for Innovation in Imaging Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Eman Akam
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Shadi A Esfahani
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Lida P Hariri
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicholas J Rotile
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
- The Institute for Innovation in Imaging Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Elizabeth Crowley
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Sydney B Montesi
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Grae Arabasz
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ciprian Catana
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
- The Institute for Innovation in Imaging Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Florian J Fintelmann
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Peter Caravan
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
- The Institute for Innovation in Imaging Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michael Lanuti
- Division of Thoracic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
13
|
Sharon S, Daher-Ghanem N, Zaid D, Gough MJ, Kravchenko-Balasha N. The immunogenic radiation and new players in immunotherapy and targeted therapy for head and neck cancer. FRONTIERS IN ORAL HEALTH 2023; 4:1180869. [PMID: 37496754 PMCID: PMC10366623 DOI: 10.3389/froh.2023.1180869] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 06/27/2023] [Indexed: 07/28/2023] Open
Abstract
Although treatment modalities for head and neck cancer have evolved considerably over the past decades, survival rates have plateaued. The treatment options remained limited to definitive surgery, surgery followed by fractionated radiotherapy with optional chemotherapy, and a definitive combination of fractionated radiotherapy and chemotherapy. Lately, immunotherapy has been introduced as the fourth modality of treatment, mainly administered as a single checkpoint inhibitor for recurrent or metastatic disease. While other regimens and combinations of immunotherapy and targeted therapy are being tested in clinical trials, adapting the appropriate regimens to patients and predicting their outcomes have yet to reach the clinical setting. Radiotherapy is mainly regarded as a means to target cancer cells while minimizing the unwanted peripheral effect. Radiotherapy regimens and fractionation are designed to serve this purpose, while the systemic effect of radiation on the immune response is rarely considered a factor while designing treatment. To bridge this gap, this review will highlight the effect of radiotherapy on the tumor microenvironment locally, and the immune response systemically. We will review the methodology to identify potential targets for therapy in the tumor microenvironment and the scientific basis for combining targeted therapy and radiotherapy. We will describe a current experience in preclinical models to test these combinations and propose how challenges in this realm may be faced. We will review new players in targeted therapy and their utilization to drive immunogenic response against head and neck cancer. We will outline the factors contributing to head and neck cancer heterogeneity and their effect on the response to radiotherapy. We will review in-silico methods to decipher intertumoral and intratumoral heterogeneity and how these algorithms can predict treatment outcomes. We propose that (a) the sequence of surgery, radiotherapy, chemotherapy, and targeted therapy should be designed not only to annul cancer directly, but to prime the immune response. (b) Fractionation of radiotherapy and the extent of the irradiated field should facilitate systemic immunity to develop. (c) New players in targeted therapy should be evaluated in translational studies toward clinical trials. (d) Head and neck cancer treatment should be personalized according to patients and tumor-specific factors.
Collapse
Affiliation(s)
- Shay Sharon
- Department of Oral and Maxillofacial Surgery, Hadassah Medical Center, Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Oral and Maxillofacial Surgery, Boston University and Boston Medical Center, Boston, MA, United States
| | - Narmeen Daher-Ghanem
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Deema Zaid
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michael J. Gough
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, United States
| | - Nataly Kravchenko-Balasha
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
14
|
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
|
15
|
Hanson I, Pitman KE, Edin NFJ. The Role of TGF-β3 in Radiation Response. Int J Mol Sci 2023; 24:ijms24087614. [PMID: 37108775 PMCID: PMC10141893 DOI: 10.3390/ijms24087614] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
Transforming growth factor-beta 3 (TGF-β3) is a ubiquitously expressed multifunctional cytokine involved in a range of physiological and pathological conditions, including embryogenesis, cell cycle regulation, immunoregulation, and fibrogenesis. The cytotoxic effects of ionizing radiation are employed in cancer radiotherapy, but its actions also influence cellular signaling pathways, including that of TGF-β3. Furthermore, the cell cycle regulating and anti-fibrotic effects of TGF-β3 have identified it as a potential mitigator of radiation- and chemotherapy-induced toxicity in healthy tissue. This review discusses the radiobiology of TGF-β3, its induction in tissue by ionizing radiation, and its potential radioprotective and anti-fibrotic effects.
Collapse
Affiliation(s)
- Ingunn Hanson
- Department of Physics, University of Oslo, 0371 Oslo, Norway
| | | | - Nina F J Edin
- Department of Physics, University of Oslo, 0371 Oslo, Norway
| |
Collapse
|
16
|
Fish BL, Hart B, Gasperetti T, Narayanan J, Gao F, Veley D, Pierce L, Himburg HA, MacVittie T, Medhora M. IPW-5371 mitigates the delayed effects of acute radiation exposure in WAG/RijCmcr rats when started 15 days after PBI with bone marrow sparing. Int J Radiat Biol 2023; 99:1119-1129. [PMID: 36794325 PMCID: PMC10330589 DOI: 10.1080/09553002.2023.2173825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/09/2023] [Accepted: 01/16/2023] [Indexed: 02/17/2023]
Abstract
PURPOSE To test IPW-5371 for the mitigation of the delayed effects of acute radiation exposure (DEARE). Survivors of acute radiation exposure are at risk for developing delayed multi-organ toxicities; however, there are no FDA-approved medical countermeasures (MCM) to mitigate DEARE. METHODS WAG/RijCmcr female rat model of partial-body irradiation (PBI), by shielding part of one hind leg, was used to test IPW-5371 (7 and 20 mg kg-1 d-1) for mitigation of lung and kidney DEARE when started 15 d after PBI. Rats were fed known amounts of IPW-5371 using a syringe, instead of delivery by daily oral gavage, sparing exacerbation of esophageal injury by radiation. The primary endpoint, all-cause morbidity was assessed over 215 d. Secondary endpoints: body weight, breathing rate and blood urea nitrogen were also assessed. RESULTS IPW-5371 enhanced survival (primary endpoint) as well as attenuated secondary endpoints of lung and kidney injuries by radiation. CONCLUSION To provide a window for dosimetry and triage, as well as avoid oral delivery during the acute radiation syndrome (ARS), the drug regimen was started at 15 d after 13.5 Gy PBI. The experimental design to test mitigation of DEARE was customized for translation in humans, using an animal model of radiation that was designed to simulate a radiologic attack or accident. The results support advanced development of IPW-5371 to mitigate lethal lung and kidney injuries after irradiation of multiple organs.
Collapse
Affiliation(s)
- Brian L. Fish
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226
| | - Barry Hart
- Innovation Pathways, Palo Alto, CA, 94301
| | - Tracy Gasperetti
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226
| | - Jayashree Narayanan
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226
| | - Feng Gao
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226
| | - Dana Veley
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226
| | - Lauren Pierce
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226
| | - Heather A. Himburg
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226
| | - Thomas MacVittie
- Innovation Pathways, Palo Alto, CA, 94301
- Department of Radiation Oncology, University of Maryland, School of Medicine, Baltimore, MD 21201
| | - Meetha Medhora
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, 53226
| |
Collapse
|
17
|
Sarkar M, Nguyen T, Gundre E, Ogunlusi O, El-Sobky M, Giri B, Sarkar TR. Cancer-associated fibroblasts: The chief architect in the tumor microenvironment. Front Cell Dev Biol 2023; 11:1089068. [PMID: 36793444 PMCID: PMC9923123 DOI: 10.3389/fcell.2023.1089068] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/12/2023] [Indexed: 01/31/2023] Open
Abstract
Stromal heterogeneity of tumor microenvironment (TME) plays a crucial role in malignancy and therapeutic resistance. Cancer-associated fibroblasts (CAFs) are one of the major players in tumor stroma. The heterogeneous sources of origin and subsequent impacts of crosstalk with breast cancer cells flaunt serious challenges before current therapies to cure triple-negative breast cancer (TNBC) and other cancers. The positive and reciprocal feedback of CAFs to induce cancer cells dictates their mutual synergy in establishing malignancy. Their substantial role in creating a tumor-promoting niche has reduced the efficacy of several anti-cancer treatments, including radiation, chemotherapy, immunotherapy, and endocrine therapy. Over the years, there has been an emphasis on understanding CAF-induced therapeutic resistance in order to enhance cancer therapy results. CAFs, in the majority of cases, employ crosstalk, stromal management, and other strategies to generate resilience in surrounding tumor cells. This emphasizes the significance of developing novel strategies that target particular tumor-promoting CAF subpopulations, which will improve treatment sensitivity and impede tumor growth. In this review, we discuss the current understanding of the origin and heterogeneity of CAFs, their role in tumor progression, and altering the tumor response to therapeutic agents in breast cancer. In addition, we also discuss the potential and possible approaches for CAF-mediated therapies.
Collapse
Affiliation(s)
- Mrinmoy Sarkar
- Department of Biology, Texas A&M University, College Station, TX, United States
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Tristan Nguyen
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Esheksha Gundre
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Olajumoke Ogunlusi
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Mohanad El-Sobky
- Department of Biology, Texas A&M University, College Station, TX, United States
| | - Biplab Giri
- Department of Physiology, University of Gour Banga, English Bazar, India
| | - Tapasree Roy Sarkar
- Department of Biology, Texas A&M University, College Station, TX, United States
| |
Collapse
|
18
|
The Normal, the Radiosensitive, and the Ataxic in the Era of Precision Radiotherapy: A Narrative Review. Cancers (Basel) 2022; 14:cancers14246252. [PMID: 36551737 PMCID: PMC9776433 DOI: 10.3390/cancers14246252] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
(1) Background: radiotherapy is a cornerstone of cancer treatment. When delivering a tumoricidal dose, the risk of severe late toxicities is usually kept below 5% using dose-volume constraints. However, individual radiation sensitivity (iRS) is responsible (with other technical factors) for unexpected toxicities after exposure to a dose that induces no toxicity in the general population. Diagnosing iRS before radiotherapy could avoid unnecessary toxicities in patients with a grossly normal phenotype. Thus, we reviewed iRS diagnostic data and their impact on decision-making processes and the RT workflow; (2) Methods: following a description of radiation toxicities, we conducted a critical review of the current state of the knowledge on individual determinants of cellular/tissue radiation; (3) Results: tremendous advances in technology now allow minimally-invasive genomic, epigenetic and functional testing and a better understanding of iRS. Ongoing large translational studies implement various tests and enriched NTCP models designed to improve the prediction of toxicities. iRS testing could better support informed radiotherapy decisions for individuals with a normal phenotype who experience unusual toxicities. Ethics of medical decisions with an accurate prediction of personalized radiotherapy's risk/benefits and its health economics impact are at stake; (4) Conclusions: iRS testing represents a critical unmet need to design personalized radiotherapy protocols relying on extended NTCP models integrating iRS.
Collapse
|
19
|
Shaghaghi Z, Alvandi M, Farzipour S, Dehbanpour MR, Nosrati S. A review of effects of atorvastatin in cancer therapy. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 40:27. [PMID: 36459301 DOI: 10.1007/s12032-022-01892-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 11/08/2022] [Indexed: 12/03/2022]
Abstract
Cancer is one of the most challenging diseases to manage. A sizeable number of researches are done each year to find better diagnostic and therapeutic strategies. At the present time, a package of chemotherapy, targeted therapy, radiotherapy, and immunotherapy is available to cope with cancer cells. Regarding chemo-radiation therapy, low effectiveness and normal tissue toxicity are like barriers against optimal response. To remedy the situation, some agents have been proposed as adjuvants to improve tumor responses. Statins, the known substances for reducing lipid, have shown a considerable capability for cancer treatment. Among them, atorvastatin as a reductase (HMG-CoA) inhibitor might affect proliferation, migration, and survival of cancer cells. Since finding an appropriate adjutant is of great importance, numerous studies have been conducted to precisely unveil antitumor effects of atorvastatin and its associated pathways. In this review, we aim to comprehensively review the most highlighted studies which focus on the use of atorvastatin in cancer therapy.
Collapse
Affiliation(s)
- Zahra Shaghaghi
- Cancer Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.,Cardiovascular Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maryam Alvandi
- Cardiovascular Research Center, Hamadan University of Medical Sciences, Hamadan, Iran. .,Department of Nuclear Medicine and Molecular Imaging, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Soghra Farzipour
- Department of Cardiology, Cardiovascular Diseases Research Center, School of Medicine, Heshmat Hospital, Guilan University of Medical Sciences, Rasht, Iran.,Department of Pharmaceutical Biotechnology, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Reza Dehbanpour
- Department of Radiology, School of Paramedicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sahar Nosrati
- Institute of Nuclear Chemistry and Technology, Dorodna 16 Str, 03-195, Warsaw, Poland
| |
Collapse
|
20
|
A role for endothelial alpha-mannosidase MAN1C1 in radiation-induced immune cell recruitment. iScience 2022; 25:105482. [DOI: 10.1016/j.isci.2022.105482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 10/06/2022] [Accepted: 10/28/2022] [Indexed: 11/15/2022] Open
|
21
|
Fatima M, Karwasra R, Almalki WH, Sahebkar A, Kesharwani P. Galactose engineered nanocarriers: Hopes and hypes in cancer therapy. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2022.111759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
22
|
Wei F, Neal CJ, Sakthivel TS, Fu Y, Omer M, Adhikary A, Ward S, Ta KM, Moxon S, Molinari M, Asiatico J, Kinzel M, Yarmolenko SN, San Cheong V, Orlovskaya N, Ghosh R, Seal S, Coathup M. A novel approach for the prevention of ionizing radiation-induced bone loss using a designer multifunctional cerium oxide nanozyme. Bioact Mater 2022; 21:547-565. [PMID: 36185749 PMCID: PMC9507991 DOI: 10.1016/j.bioactmat.2022.09.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/18/2022] Open
Abstract
The disability, mortality and costs due to ionizing radiation (IR)-induced osteoporotic bone fractures are substantial and no effective therapy exists. Ionizing radiation increases cellular oxidative damage, causing an imbalance in bone turnover that is primarily driven via heightened activity of the bone-resorbing osteoclast. We demonstrate that rats exposed to sublethal levels of IR develop fragile, osteoporotic bone. At reactive surface sites, cerium ions have the ability to easily undergo redox cycling: drastically adjusting their electronic configurations and versatile catalytic activities. These properties make cerium oxide nanomaterials fascinating. We show that an engineered artificial nanozyme composed of cerium oxide, and designed to possess a higher fraction of trivalent (Ce3+) surface sites, mitigates the IR-induced loss in bone area, bone architecture, and strength. These investigations also demonstrate that our nanozyme furnishes several mechanistic avenues of protection and selectively targets highly damaging reactive oxygen species, protecting the rats against IR-induced DNA damage, cellular senescence, and elevated osteoclastic activity in vitro and in vivo. Further, we reveal that our nanozyme is a previously unreported key regulator of osteoclast formation derived from macrophages while also directly targeting bone progenitor cells, favoring new bone formation despite its exposure to harmful levels of IR in vitro. These findings open a new approach for the specific prevention of IR-induced bone loss using synthesis-mediated designer multifunctional nanomaterials.
Collapse
Key Words
- ALP, Alkaline phosphatase
- BMSC, Bone marrow derived mesenchymal stem cells
- Bone resorption
- Bone strength
- CAT, Catalase
- COLI, Collagen type I
- CTSK, Cathepsin K
- CTX-1, Cross-linked C-telopeptide of type I collagen
- CeONPs, Cerium oxide nanoparticles
- Cerium oxide
- DFT, Density functional theory
- DNA, Deoxyribonucleic acid
- EPR, Electron paramagnetic resonance
- FDA, Food and Drug Administration
- GPX, Glutathione peroxidase
- Gy, Gray
- HIF1α, Hypoxia-inducible factor 1 alpha
- IL-1β, Interleukin 1 beta
- IL-6, Interleukin 6
- IR, Ionizing radiation
- Ionizing radiation
- MNGC, Multinucleated giant cell
- Nanozyme
- OCN, Osteocalcin
- Osteoporosis
- RANKL, Receptor activator of nuclear factor kappa-Β ligand
- ROS, Reactive oxygen species
- SAED, Selected area electron diffraction
- SOD, Superoxide dismutase
- TRAP, Tartrate-resistant acid phosphatase
- XPS, X-ray photoelectron spectroscopy
Collapse
Affiliation(s)
- Fei Wei
- Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Craig J. Neal
- Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC), Materials Science and Engineering, College of Medicine, University of Central Florida, Orlando, FL, USA
| | | | - Yifei Fu
- Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC), Materials Science and Engineering, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Mahmoud Omer
- Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Amitava Adhikary
- Department of Chemistry, Oakland University, Rochester, MI, MI, USA
| | - Samuel Ward
- Department of Chemistry, Oakland University, Rochester, MI, MI, USA
| | - Khoa Minh Ta
- School of Applied Sciences, Department of Chemical Sciences, University of Huddersfield, UK
| | - Samuel Moxon
- School of Applied Sciences, Department of Chemical Sciences, University of Huddersfield, UK
| | - Marco Molinari
- School of Applied Sciences, Department of Chemical Sciences, University of Huddersfield, UK
| | - Jackson Asiatico
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, USA
| | - Michael Kinzel
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, USA
| | - Sergey N. Yarmolenko
- Engineering Research Center for Revolutionizing Biomaterials, North Carolina A & T University, Greensboro, NC, USA
| | - Vee San Cheong
- Department of Automatic Control and Systems Engineering, Insigneo Institute for In Silico Medicine, University of Sheffield, Sheffield, S1 3JD, UK
| | - Nina Orlovskaya
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, USA
| | - Ranajay Ghosh
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, FL, USA
| | - Sudipta Seal
- Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA
- Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC), Materials Science and Engineering, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Melanie Coathup
- Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA
- Corresponding author. Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, USA.
| |
Collapse
|
23
|
Isoyama S, Yamaguchi K, Imano N, Sakamoto S, Horimasu Y, Masuda T, Miyamoto S, Nakashima T, Iwamoto H, Fujitaka K, Hamada H, Nagata Y, Hattori N. Predictive role of circulatory levels of high-mobility group box 1 for radiation pneumonitis in patients with non-small cell lung cancer treated with definitive thoracic radiotherapy. Int J Clin Oncol 2022; 27:1698-1705. [PMID: 36057047 DOI: 10.1007/s10147-022-02239-0] [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: 03/15/2022] [Accepted: 08/11/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND High-mobility group box 1 (HMGB1) is a pro-inflammatory protein associated with the pathophysiology of lung injury and lung tumorigenesis. Here, we investigated the predictive potential of serum HMGB1 levels for radiation pneumonitis in patients with lung cancer. METHODS This was a retrospective biomarker study of 73 patients with non-small cell lung cancer treated with definitive thoracic radiotherapy between August 2007 and January 2021. We measured HMGB1 levels in serum stored before treatment, and analyzed its association with the development of grade ≥ 2 or grade ≥ 3 radiation pneumonitis. Additionally, baseline characteristics affecting HMGB1 levels were identified. RESULTS Of the 73 patients, 21 (28.8%) and 6 (8.2%) patients experienced grade 2 and ≥ 3 radiation pneumonitis, respectively. Univariate and multivariate logistic regression analyses revealed that higher baseline levels of serum HMGB1 were significantly associated with a higher risk of grade ≥ 3, but not grade ≥ 2, radiation pneumonitis. The incidence of grade ≥ 3 radiation pneumonitis was higher in patients with HMGB1 levels ≥ 6.2 ng/mL than in those with levels < 6.2 ng/mL (25.0% vs. 3.5%, p = 0.019). Baseline serum levels of HMGB1 were independently and positively associated with gross tumor volume. CONCLUSIONS Higher serum HMGB1 levels were significantly associated with the risk of grade ≥ 3 radiation pneumonitis in patients with lung cancer, and therefore, HMGB1 could be a potential blood biomarker for predicting severe radiation pneumonitis.
Collapse
Affiliation(s)
- Shoko Isoyama
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Kakuhiro Yamaguchi
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| | - Nobuki Imano
- Department of Radiation Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shinjiro Sakamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Yasushi Horimasu
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Takeshi Masuda
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Shintaro Miyamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Taku Nakashima
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Hiroshi Iwamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Kazunori Fujitaka
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Hironobu Hamada
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Yasushi Nagata
- Department of Radiation Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Noboru Hattori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
24
|
Abd Elmonem HA, Mater SN, Eldeighdye SM. Protective role of vitamin D against radiation hazards in rats fed on high fat diet. JOURNAL OF RADIATION RESEARCH AND APPLIED SCIENCES 2022. [DOI: 10.1016/j.jrras.2022.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
25
|
Fu X, Li T, Yao Q. The Effect of Ophiopogonin C in Ameliorating Radiation-Induced Pulmonary Fibrosis in C57BL/6 Mice: An Update Study. Front Oncol 2022; 12:811183. [PMID: 35433490 PMCID: PMC9007236 DOI: 10.3389/fonc.2022.811183] [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: 11/08/2021] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Background The aim of this study was to assess and update the protective effects and underlying mechanisms of Ophiopogonin C (OP-C), a biologically active component separated and purified from Ophiopogon japonicus, in ameliorating radiation-induced pulmonary fibrosis in C57BL/6 mice administered thoracic radiation. Methods and Materials We randomly divided 75 mice into five groups and administered a dose of 12-Gy whole thoracic radiation to establish a pulmonary fibrosis animal model. Mice were treated with OP-C or dexamethasone combined with or without cephalexin by daily gavage for 4 weeks. All mice were sacrificed after the completion of thoracic irradiation at 28 weeks. Serum levels of interleukin-6 and transforming growth factor-β1 (TGF-β1) were evaluated. Moreover, superoxide dismutase (SOD) levels in lung tissue were measured. The severity of fibrosis was evaluated using the hydroxyproline content of the lung tissue. The pathological changes in the five groups were detected by hematoxylin and eosin and Masson trichrome staining. Smooth muscle actin expression was detected using immunohistochemical staining. Matrix metalloproteinases-2 (MMP-2) and tissue inhibitors of metalloproteases-2 (TIMP-2) were examined by immunohistochemical staining of the lung sections, and semiquantitative analysis was used to calculate the expression of MMP-2 and TIMP-2. Results Irradiated mice treated with OP-C or DXE combined with or without cephalexin significantly reduced mortality in mice and fibrosis levels by 1) reducing the deposition of collagen and accumulation of inflammatory cells and fibroblasts, 2) downgrading levels of the promote-fibrosis cytokine TGF-β1, and 3) increasing SOD activity in the lung tissue compared with that of irradiated mice without treatment. However, there were no statistical differences in fibrosis levels among the irradiated mice treated with OP-C or DXE combined with or without cephalexin. Conclusion OP-C significantly ameliorates radiation-induced pulmonary fibrosis and may be a promising therapeutic strategy for this disorder.
Collapse
Affiliation(s)
- Xiaobin Fu
- Department of Radiation Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Tingting Li
- Department of Radiation Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Qiwei Yao
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou, China
| |
Collapse
|
26
|
Arora A, Bhuria V, Singh S, Pathak U, Mathur S, Hazari PP, Roy BG, Sandhir R, Soni R, Dwarakanath BS, Bhatt AN. Amifostine analog, DRDE-30, alleviates radiation induced lung damage by attenuating inflammation and fibrosis. Life Sci 2022; 298:120518. [PMID: 35367468 DOI: 10.1016/j.lfs.2022.120518] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/18/2022] [Accepted: 03/26/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Radiotherapy of thoracic neoplasms and accidental radiation exposure often results in pneumonitis and fibrosis of lungs. Here, we investigated the potential of amifostine analogs: DRDE-07, DRDE-30, and DRDE-35, in alleviating radiation-induced lung damage. METHODS C57BL/6 mice were exposed to 13.5 Gy thoracic irradiation, 30 min after intraperitoneal administration of the analogs, and assessed for modulation of the pathological response at 12 and 24 weeks. KEY FINDINGS DRDE-07, DRDE-30 and DRDE-35 increased the survival of irradiated mice from 20% to 30%, 80% and 70% respectively. Reduced parenchymal opacity (X-ray CT) in the lungs of DRDE-30 pre-treated mice corroborated well with the significant decrease in Ashcroft score (p < 0.01). Two-fold increase in SOD and catalase activities (p < 0.05), coupled with a 50% increase in GSH content and a 60% decrease in MDA content (p < 0.05) suggested restoration of the antioxidant defence system. A 20% to 40% decrease in radiation-induced apoptotic and mitotic death in the lung tissue (micronuclei: p < 0.01), resulted in attenuated lung and vascular permeability (FITC-Dextran leakage) by 50% (p < 0.01), and a commensurate reduction (~50%) in leukocyte infiltration in the injured tissue (p < 0.05). DRDE-30 abrogated the activation of pro-inflammatory NF-κB and p38/MAPK signaling cascades, suppressing the release of pro-inflammatory cytokines (IL-1β: p < 0.05; TNF-α: p < 0.05; IL-6: p < 0.05) and up-regulation of CAMs on the endothelial cell surface. Reduction in hydroxyproline content (p < 0.01) and collagen suggested inhibition of lung fibrosis which was associated with attenuation of TGF-β/Smad pathway-mediated-EMT. CONCLUSION DRDE-30 could be a potential prophylactic agent against radiation-induced lung injury.
Collapse
Affiliation(s)
- Aastha Arora
- Institute of Nuclear Medicine & Allied Sciences, Delhi, India; Department of Biochemistry, Panjab University, Chandigarh, India
| | - Vikas Bhuria
- Institute of Nuclear Medicine & Allied Sciences, Delhi, India
| | - Saurabh Singh
- Institute of Nuclear Medicine & Allied Sciences, Delhi, India
| | - Uma Pathak
- Defence Research and Development Establishment, Gwalior, India
| | - Sweta Mathur
- Defence Research and Development Establishment, Gwalior, India
| | - Puja P Hazari
- Institute of Nuclear Medicine & Allied Sciences, Delhi, India
| | - Bal G Roy
- Institute of Nuclear Medicine & Allied Sciences, Delhi, India
| | - Rajat Sandhir
- Department of Biochemistry, Panjab University, Chandigarh, India
| | - Ravi Soni
- Institute of Nuclear Medicine & Allied Sciences, Delhi, India
| | - Bilikere S Dwarakanath
- Institute of Nuclear Medicine & Allied Sciences, Delhi, India; Central Research Facility, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | | |
Collapse
|
27
|
Tanigami Y, Kawai Y, Kaba S, Uozumi R, Ohnishi H, Kita T, Omori K, Kishimoto Y. Establishment of a radiation-induced vocal fold fibrosis mouse model. Biochem Biophys Res Commun 2022; 601:31-37. [DOI: 10.1016/j.bbrc.2022.02.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 02/18/2022] [Indexed: 11/26/2022]
|
28
|
Cytlak UM, Dyer DP, Honeychurch J, Williams KJ, Travis MA, Illidge TM. Immunomodulation by radiotherapy in tumour control and normal tissue toxicity. Nat Rev Immunol 2022; 22:124-138. [PMID: 34211187 DOI: 10.1038/s41577-021-00568-1] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2021] [Indexed: 12/12/2022]
Abstract
Radiotherapy (RT) is a highly effective anticancer treatment that is delivered to more than half of all patients with cancer. In addition to the well-documented direct cytotoxic effects, RT can have immunomodulatory effects on the tumour and surrounding tissues. These effects are thought to underlie the so-called abscopal responses, whereby RT generates systemic antitumour immunity outside the irradiated tumour. The full scope of these immune changes remains unclear but is likely to involve multiple components, such as immune cells, the extracellular matrix, endothelial and epithelial cells and a myriad of chemokines and cytokines, including transforming growth factor-β (TGFβ). In normal tissues exposed to RT during cancer therapy, acute immune changes may ultimately lead to chronic inflammation and RT-induced toxicity and organ dysfunction, which limits the quality of life of survivors of cancer. Here we discuss the emerging understanding of RT-induced immune effects with particular focus on the lungs and gut and the potential immune crosstalk that occurs between these tissues.
Collapse
Affiliation(s)
- Urszula M Cytlak
- Lydia Becker Institute for Immunology and Inflammation, Wellcome Centre for Cell-Matrix Research, Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
- Targeted Therapy Group, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| | - Douglas P Dyer
- Lydia Becker Institute for Immunology and Inflammation, Wellcome Centre for Cell-Matrix Research, Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jamie Honeychurch
- Targeted Therapy Group, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Kaye J Williams
- Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Mark A Travis
- Lydia Becker Institute for Immunology and Inflammation, Wellcome Centre for Cell-Matrix Research, Manchester Collaborative Centre for Inflammation Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| | - Timothy M Illidge
- Targeted Therapy Group, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
29
|
Dai S, Wen Y, Luo P, Ma L, Liu Y, Ai J, Shi C. Therapeutic implications of exosomes in the treatment of radiation injury. BURNS & TRAUMA 2022; 10:tkab043. [PMID: 35071650 PMCID: PMC8778593 DOI: 10.1093/burnst/tkab043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 11/11/2021] [Indexed: 12/28/2022]
Abstract
Radiotherapy is one of the main cancer treatments, but it may damage normal tissue and cause various side effects. At present, radioprotective agents used in clinics have side effects such as nausea, vomiting, diarrhea and hypotension, which limit their clinical application. It has been found that exosomes play an indispensable role in radiation injury. Exosomes are lipid bilayer vesicles that carry various bioactive substances, such as proteins, lipids and microRNA (miRNA), that play a key role in cell-to-cell communication and affect tissue injury and repair. In addition, studies have shown that radiation can increase the uptake of exosomes in cells and affect the composition and secretion of exosomes. Here, we review the existing studies and discuss the effects of radiation on exosomes and the role of exosomes in radiation injury, aiming to provide new insights for the treatment of radiation injury.
Collapse
Affiliation(s)
| | | | | | | | | | - Junhua Ai
- Correspondence. Junhua Ai, ; Chunmeng Shi,
| | | |
Collapse
|
30
|
Feasibility study for inducing the skeletal muscle fibrosis via irradiation using two mouse strains. Jpn J Radiol 2021; 40:466-475. [PMID: 34841459 DOI: 10.1007/s11604-021-01219-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 11/05/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE Although the mechanism of onset and progression of radiation-induced fibrosis (RIF) has been studied, most studies to date have focused on pulmonary fibrosis. There are few studies on murine RIF in the skeletal muscle, and the pathogenic mechanism remains unclear. This pilot study aimed to evaluate the feasibility to create a murine model of RIF in the skeletal muscle and analyze strain differences in fibrosis sensitivity. MATERIALS AND METHODS Two mouse strains, C57BL/6 and C3H/He, were used. Their right hind limbs were irradiated at a dose of 25 Gy once a week for three fractions. Gastrocnemius muscles were collected at day 4, and weeks 2, 4, 8, 12, and 24 after the third irradiation and subjected to histopathological examination and immunoblotting. RESULTS In C57BL/6 mice, chronic inflammation and an increased expression of transforming growth factor-β (TGF-β) and fibronectin were observed 2 weeks after irradiation. A significant increase in fibrosis was detected after 8 weeks. However, in C3H/He mice, the expression of TGF-β and fibronectin increased 8 weeks after irradiation, and fibrosis significantly increased after 12 weeks. Moreover, the degrees of inflammation and fibrosis were more remarkable in C57BL/6 mice than in C3H/He mice. CONCLUSION The onset and degree of fibrosis may be associated with the expression of TGF-β and fibronectin, and inflammation, in a strain-specific manner. Therefore, a murine model of RIF in the skeletal muscle could be created using the indicated method, suggesting that the C57BL/6 strain is more sensitive to fibrosis in the skeletal muscle, as well as the lung, than the C3H/He strain. Radiation-induced fibrosis in the skeletal muscle could be detected in C57BL/6 and C3H/He mice, with C57BL/6 mice being more sensitive to fibrosis in the skeletal muscle than C3H/He mice.
Collapse
|
31
|
Hellevik T, Berzaghi R, Lode K, Islam A, Martinez-Zubiaurre I. Immunobiology of cancer-associated fibroblasts in the context of radiotherapy. J Transl Med 2021; 19:437. [PMID: 34663337 PMCID: PMC8524905 DOI: 10.1186/s12967-021-03112-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 10/08/2021] [Indexed: 12/14/2022] Open
Abstract
Radiotherapy (RT) still represents a mainstay of treatment in clinical oncology. Traditionally, the effectiveness of radiotherapy has been attributed to the killing potential of ionizing radiation (IR) over malignant cells, however, it has become clear that therapeutic efficacy of RT also involves activation of innate and adaptive anti-tumor immune responses. Therapeutic irradiation of the tumor microenvironment (TME) provokes profound cellular and biological reconfigurations which ultimately may influence immune recognition. As one of the major constituents of the TME, cancer-associated fibroblasts (CAFs) play central roles in cancer development at all stages and are recognized contributors of tumor immune evasion. While some studies argue that RT affects CAFs negatively through growth arrest and impaired motility, others claim that exposure of fibroblasts to RT promotes their conversion into a more activated phenotype. Nevertheless, despite the well-described immunoregulatory functions assigned to CAFs, little is known about the interplay between CAFs and immune cells in the context of RT. In this review, we go over current literature on the effects of radiation on CAFs and the influence that CAFs have on radiotherapy outcomes, and we summarize present knowledge on the transformed cellular crosstalk between CAFs and immune cells after radiation.
Collapse
Affiliation(s)
- Turid Hellevik
- Department of Radiation Oncology, University Hospital of Northern Norway, Tromsø, Norway
| | - Rodrigo Berzaghi
- Department of Clinical Medicine, Faculty of Health Sciences, UiT-the Arctic University of Norway, Tromsø, Norway
| | - Kristin Lode
- Department of Clinical Medicine, Faculty of Health Sciences, UiT-the Arctic University of Norway, Tromsø, Norway
| | - Ashraful Islam
- Department of Clinical Medicine, Faculty of Health Sciences, UiT-the Arctic University of Norway, Tromsø, Norway
| | - Inigo Martinez-Zubiaurre
- Department of Clinical Medicine, Faculty of Health Sciences, UiT-the Arctic University of Norway, Tromsø, Norway.
| |
Collapse
|
32
|
Therapeutic approach of adipose-derived mesenchymal stem cells in refractory peptic ulcer. Stem Cell Res Ther 2021; 12:515. [PMID: 34565461 PMCID: PMC8474857 DOI: 10.1186/s13287-021-02584-3] [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: 06/25/2021] [Accepted: 09/04/2021] [Indexed: 12/14/2022] Open
Abstract
Peptic ulcer is one of the most common gastrointestinal tract disorders worldwide, associated with challenges such as refractory morbidity, bleeding, interference with use of anticoagulants, and potential side effects associated with long-term use of proton pump inhibitors. A peptic ulcer is a defect in gastric or duodenal mucosa extending from muscularis mucosa to deeper layers of the stomach wall. In most cases, ulcers respond to standard treatments. However, in some people, peptic ulcer becomes resistant to conventional treatment or recurs after initially successful therapy. Therefore, new and safe treatments, including the use of stem cells, are highly favored for these patients. Adipose-derived mesenchymal stem cells are readily available in large quantities with minimal invasive intervention, and isolation of adipose-derived mesenchymal stromal stem cells (ASC) produces large amounts of stem cells, which are essential for cell-based and restorative therapies. These cells have high flexibility and can differentiate into several types of cells in vitro. This article will investigate the effects and possible mechanisms and signaling pathways of adipose tissue-derived mesenchymal stem cells in patients with refractory peptic ulcers.
Collapse
|
33
|
Singh J, Thachil T, Eapen MS, Lim A, Sufyan W, Rawson R, Duncan H, De Ieso P, Sohal SS. Immunohistochemical investigation of cytokine expression levels as biomarkers in transrectal ultrasound-guided needle biopsy specimens of prostate adenocarcinoma. Mol Clin Oncol 2021; 15:191. [PMID: 34405051 DOI: 10.3892/mco.2021.2353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 06/24/2021] [Indexed: 12/27/2022] Open
Abstract
Cytokines influence the biological behaviour of prostate cancer (PC) and may influence patient outcome and serve as useful prognostic biomarkers. The aim of the present study was to evaluate cytokine expression levels in prostatic needle biopsy specimens and the association with clinicopathological characteristics of patients with PC. A total of 18 patients with PC who underwent transrectal ultrasound (TRUS) guided prostate biopsy were included in the clinical study. These patients were naïve to radiotherapy (RT) or androgen deprivation therapy prior to TRUS biopsy and clinical follow up data was collected. Cytokine expression levels were analysed by using immunohistochemistry and Spearman's correlation test was used to determine the correlation between cytokine expression and clinicopathological characteristics. Expression levels of pro-inflammatory TNF-α and IL-6 decreased as Gleason score (GS) increased; however, a statistically significant difference was not detected. A statically significant correlation was observed between needle biopsy specimen and pre-RT plasma sample expression levels of pro-inflammatory TNF-α and IL-6 (P=0.01 and P=0.05, respectively) and anti-inflammatory TGF-β1 (P=0.05). However, further studies are needed to confirm these results using a larger sample size to confirm the prognostic value of pro-inflammatory TNF-α and IL-6 and anti-inflammatory TGF-β1 in patients with PC.
Collapse
Affiliation(s)
- Jagtar Singh
- College of Health and Human Sciences, Charles Darwin University, Northern Territory 0810, Australia.,Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania 7248, Australia
| | - Thanuja Thachil
- Ballarat Austin Radiation Oncology Centre, Victoria 3350, Australia
| | - Mathew Suji Eapen
- Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania 7248, Australia
| | - Aijye Lim
- Department of Anatomical Pathology, Royal Darwin Hospital 0810, Australia
| | - Wajiha Sufyan
- Department of Anatomical Pathology, Royal Darwin Hospital 0810, Australia
| | - Robert Rawson
- Department of Anatomical Pathology, Royal Darwin Hospital 0810, Australia
| | - Henry Duncan
- Urology Department, Darwin Private Hospital, Northern Territory 0810, Australia
| | - Paolo De Ieso
- Peter MacCallum Cancer Centre, Victoria 3000, Australia
| | - Sukhwinder Singh Sohal
- Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania 7248, Australia
| |
Collapse
|
34
|
Nakamura N. Reexamining the role of tissue inflammation in radiation carcinogenesis: a hypothesis to explain an earlier onset of cancer. Int J Radiat Biol 2021; 97:1341-1351. [PMID: 34270352 DOI: 10.1080/09553002.2021.1955998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE Ionizing radiation is a well-known carcinogen, and epidemiologic efforts have been made to evaluate cancer risks following a radiation exposure. The basic approach has been to estimate increased levels of cancer mortality resulting from exposures to radiation, which is consistent with the somatic mutation theory of cancer. However, the possibility that an irradiation might cause an earlier onset of cancer has also been raised since the earliest days of animal studies. Recently, the mutation induction model has been challenged because it is unable to explain the observed dose-related parallel shift of entire mouse survival curves toward younger ages following an irradiation. This is because if it is assumed that only a fraction of the irradiated individuals are affected, the irradiated population would consist of two subpopulations with different mean lifespans, which makes the overall distribution of individual lifespans broader, and hence the slope of the survival curves shallower. To explain this parallel shift, it is necessary to assume that all individuals of a population are affected. As a result of these observations, possible mechanisms which could account for the parallel shift of mouse survival curves were sought by examining the radiation induction of various types of tissue damage which could facilitate an earlier onset of spontaneously arising cancers. CONCLUSION A proposed mechanism postulates that a radiation exposure leads to tissue inflammation which subsequently stimulates spontaneously arising cancers and allows them to appear earlier than usual. This notion is not unprecedented, and because the background incidence of cancer increases exponentially with an increase in age, a slight shift of the onset age toward younger ages may make it appear as if the risk is increased. In this scenario, a radiation exposure induces DNA damage, cell death, chromosome aberrations etc., which leads to the multi-pathway responses including activation of stromal fibroblasts, macrophages and various inflammatory factors. Such an inflamed microenvironment favors the growth of spontaneously arising tumor cells although currently, the sequential order or relative importance of the individual factors remains to be known.
Collapse
Affiliation(s)
- Nori Nakamura
- Department, of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima, Japan
| |
Collapse
|
35
|
Boerma M, Davis CM, Jackson IL, Schaue D, Williams JP. All for one, though not one for all: team players in normal tissue radiobiology. Int J Radiat Biol 2021; 98:346-366. [PMID: 34129427 PMCID: PMC8781287 DOI: 10.1080/09553002.2021.1941383] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE As part of the special issue on 'Women in Science', this review offers a perspective on past and ongoing work in the field of normal (non-cancer) tissue radiation biology, highlighting the work of many of the leading contributors to this field of research. We discuss some of the hypotheses that have guided investigations, with a focus on some of the critical organs considered dose-limiting with respect to radiation therapy, and speculate on where the field needs to go in the future. CONCLUSIONS The scope of work that makes up normal tissue radiation biology has and continues to play a pivotal role in the radiation sciences, ensuring the most effective application of radiation in imaging and therapy, as well as contributing to radiation protection efforts. However, despite the proven historical value of preclinical findings, recent decades have seen clinical practice move ahead with altered fractionation scheduling based on empirical observations, with little to no (or even negative) supporting scientific data. Given our current appreciation of the complexity of normal tissue radiation responses and their temporal variability, with tissue- and/or organ-specific mechanisms that include intra-, inter- and extracellular messaging, as well as contributions from systemic compartments, such as the immune system, the need to maintain a positive therapeutic ratio has never been more urgent. Importantly, mitigation and treatment strategies, whether for the clinic, emergency use following accidental or deliberate releases, or reducing occupational risk, will likely require multi-targeted approaches that involve both local and systemic intervention. From our personal perspective as five 'Women in Science', we would like to acknowledge and applaud the role that many female scientists have played in this field. We stand on the shoulders of those who have gone before, some of whom are fellow contributors to this special issue.
Collapse
Affiliation(s)
- Marjan Boerma
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Catherine M. Davis
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Isabel L. Jackson
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dörthe Schaue
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Jacqueline P. Williams
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
36
|
Impact of Chronic Obstruction Pulmonary Disease on Survival in Patients with Advanced Stage Lung Squamous Cell Carcinoma Undergoing Concurrent Chemoradiotherapy. Cancers (Basel) 2021; 13:cancers13133231. [PMID: 34203540 PMCID: PMC8268442 DOI: 10.3390/cancers13133231] [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: 06/20/2021] [Accepted: 06/26/2021] [Indexed: 12/28/2022] Open
Abstract
Simple Summary No data are available regarding the effect of chronic obstruction pulmonary disease (COPD) and COPD with acute exacerbation (COPDAE) on survival in patients with lung squamous cell carcinoma (SCC) receiving definitive concurrent chemoradiotherapy (CCRT). This study is the first to examine the survival impact of COPD in patients with lung SCC receiving definitive CCRT. COPD and its severity are significant independent risk factors for all-cause mortality in patients with stage IIIA–IIIB lung SCC receiving definitive CCRT. Hospitalization for COPDAE within 1 year before CCRT is the significant independent risk factor for lung cancer death in the patients with stage IIIA–IIIB lung SCC receiving definitive CCRT. Abstract Background: To date, no data are available regarding the effect of chronic obstruction pulmonary disease (COPD) and COPD with acute exacerbation (COPDAE) on survival in patients with lung squamous cell carcinoma (SCC) receiving definitive concurrent chemoradiotherapy (CCRT). Patients and methods: We enrolled 3986 patients with clinical stage IIIA–IIIB, unresectable lung SCC, who had received standard definitive CCRT, and categorized them into two groups based on their COPD status to compare overall survival outcomes. We also examined the effects of COPD severity (0, 1, or ≥2 hospitalizations for COPDA within 1 year before CCRT). Results: In the inverse probability of treatment weighting (IPTW)-adjusted model, the adjusted hazard ratio (aHR) (95% confidence interval (CI)) of all-cause death for COPD was 1.04 (1.01, 1.16), compared no COPD in patients with stage IIIA–IIIB lung SCC receiving definitive CCRT. In the IPTW-adjusted model, the aHRs (95% CIs) of 1 and ≥ 2 hospitalizations for COPDAE within 1 year before CCRT were 1.32 (1.19, 1.46) and 1.81 (1.49, 2.19) respectively, compared with no hospitalization for COPDAE. Conclusion: COPD and its severity are significant independent risk factors for all-cause death in patients with stage IIIA–IIIB lung SCC receiving definitive CCRT. Hospitalization for COPDAE within 1 year before CCRT is the significant independent risk factor for lung cancer death in the patients with stage IIIA–IIIB lung SCC receiving definitive CCRT.
Collapse
|
37
|
Owen DR, Sun Y, Boonstra PS, McFarlane M, Viglianti BL, Balter JM, El Naqa I, Schipper MJ, Schonewolf CA, Ten Haken RK, Kong FMS, Jolly S, Matuszak MM. Investigating the SPECT Dose-Function Metrics Associated With Radiation-Induced Lung Toxicity Risk in Patients With Non-small Cell Lung Cancer Undergoing Radiation Therapy. Adv Radiat Oncol 2021; 6:100666. [PMID: 33817412 PMCID: PMC8010578 DOI: 10.1016/j.adro.2021.100666] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 01/22/2021] [Indexed: 12/14/2022] Open
Abstract
Purpose Dose to normal lung has commonly been linked with radiation-induced lung toxicity (RILT) risk, but incorporating functional lung metrics in treatment planning may help further optimize dose delivery and reduce RILT incidence. The purpose of this study was to investigate the impact of the dose delivered to functional lung regions by analyzing perfusion (Q), ventilation (V), and combined V/Q single-photon-emission computed tomography (SPECT) dose-function metrics with regard to RILT risk in patients with non-small cell lung cancer (NSCLC) patients who received radiation therapy (RT). Methods and Materials SPECT images acquired from 88 patients with locally advanced NSCLC before undergoing conventionally fractionated RT were retrospectively analyzed. Dose was converted to the nominal dose equivalent per 2 Gy fraction, and SPECT intensities were normalized. Regional lung segments were defined, and the average dose delivered to each lung region was quantified. Three functional categorizations were defined to represent low-, normal-, and high-functioning lungs. The percent of functional lung category receiving ≥20 Gy and mean functional intensity receiving ≥20 Gy (iV20) were calculated. RILT was defined as grade 2+ radiation pneumonitis and/or clinical radiation fibrosis. A logistic regression was used to evaluate the association between dose-function metrics and risk of RILT. Results By analyzing V/Q normalized intensities and functional distributions across the population, a wide range in functional capability (especially in the ipsilateral lung) was observed in patients with NSCLC before RT. Through multivariable regression models, global lung average dose to the lower lung was found to be significantly associated with RILT, and Q and V iV20 were correlated with RILT when using ipsilateral lung metrics. Through a receiver operating characteristic analysis, combined V/Q low-function receiving ≥20 Gy (low-functioning V/Q20) in the ipsilateral lung was found to be the best predictor (area under the curce: 0.79) of RILT risk. Conclusions Irradiation of the inferior lung appears to be a locational sensitivity for RILT risk. The multivariable correlation between ipsilateral lung iV20 and RILT, as well as the association of low-functioning V/Q20 and RILT, suggest that irradiating low-functioning regions in the lung may lead to higher toxicity rates.
Collapse
Affiliation(s)
- Daniel R Owen
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Yilun Sun
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan.,Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Philip S Boonstra
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Matthew McFarlane
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Benjamin L Viglianti
- Department of Radiology, University of Michigan, Ann Arbor, Michigan.,Veterans Administration, Nuclear Medicine Service, Ann Arbor Michigan
| | - James M Balter
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Issam El Naqa
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Matthew J Schipper
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | | | - Randall K Ten Haken
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Feng-Ming S Kong
- Hong Kong University Shenzhen Hospital and Queen Mary Hospital, Hong Kong University Li Ka Shing Medical School, Department of Clinical Oncology, Hong Kong.,Department of Radiation Oncology, Case Western Reserve University, Cleveland, Ohio
| | - Shruti Jolly
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Martha M Matuszak
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
38
|
Kura B, Kalocayova B, Szeiffova Bacova B, Fulop M, Sagatova A, Sykora M, Andelova K, Abuawad Z, Slezak J. The effect of selected drugs on the mitigation of myocardial injury caused by gamma radiation. Can J Physiol Pharmacol 2021; 99:80-88. [PMID: 33438486 DOI: 10.1139/cjpp-2020-0323] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Radiation damage of healthy tissues represents one of the complications of radiotherapy effectiveness. This study is focused on the screening of potentially effective drugs routinely used in medical practice and involved in the mechanism of radiation injury, namely for radiation-induced production of free radicals in the body. Experiments in rats revealed significant reduction of oxidative stress (malondialdehyde) and inflammatory marker (tumor necrosis factor α) in 10 Gy irradiated groups after administration of atorvastatin and a slight decrease after tadalafil administration, which indicates that one of the possible mechanisms for mitigation of radiation-induced cardiac damage could be the modulation of nitric oxide (NO) in endothelium and phosphodiesterase 5. In addition, miRNAs were analyzed as potential markers and therapeutically effective molecules. Expression of miRNA-21 and miRNA-15b showed the most significant changes after irradiation. Atorvastatin and tadalafil normalized changes of miRNA (miRNA-1, miRNA-15b, miRNA-21) expression levels in irradiated hearts. This screening study concludes that administration of specific drugs could mitigate the negative impact of radiation on the heart, but more detailed experiments oriented to other aspects of drug effectiveness and their exact mechanisms are still needed.
Collapse
Affiliation(s)
- Branislav Kura
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, 841 04 Bratislava, Slovak Republic.,Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, 813 72 Bratislava, Slovak Republic
| | - Barbora Kalocayova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, 841 04 Bratislava, Slovak Republic
| | - Barbara Szeiffova Bacova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, 841 04 Bratislava, Slovak Republic
| | - Marko Fulop
- Slovak Medical University, 831 01, Bratislava, Slovak Republic
| | - Andrea Sagatova
- Faculty of Electrical Engineering and Information Technology, Institute of Nuclear and Physical Engineering, Slovak University of Technology in Bratislava, 812 19 Bratislava, Slovak Republic
| | - Matus Sykora
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, 841 04 Bratislava, Slovak Republic
| | - Katarina Andelova
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, 841 04 Bratislava, Slovak Republic
| | - Ziad Abuawad
- Faculty of Public Health, Al-Quds University, Jerusalem, Palestine
| | - Jan Slezak
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, 841 04 Bratislava, Slovak Republic
| |
Collapse
|
39
|
Aras S, Tanzer İO, Sayir N, Keleş MS, Özgeriş FB. Radiobiological comparison of flattening filter (FF) and flattening filter-free (FFF) beam in rat laryngeal tissue. Int J Radiat Biol 2021; 97:249-255. [PMID: 33320739 DOI: 10.1080/09553002.2021.1857457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE The purpose of this study is to investigate the radioprotective effect of melatonin by analyzing histopathological changes and serum biochemical levels on experimental rat models exposed to flattening filter (FF) and flattening filter-free (FFF) beam. MATERIALS AND METHODS Forty-eight healthy adult Sprague Dawley rats were randomly divided into six groups. The control (Group 1) was given no treatment, the melatonin (Group 2) was given 10 mg/kg melatonin only, the FF (Group 3) and FFF (Group 5) were given fractionated dose (Total 32 Gy, 5 consecutive days) radiotherapy only, and the FF plus melatonin (Group 4) and FFF plus melatonin (Group 6) were given 10 mg/kg melatonin 15 minutes prior to irradiation. Rats were examined for histopathology and biochemical analysis 10 days after irradiation. RESULTS When results of FF and FFF radiotherapy only groups are compared to control group, statistically significant difference in histopathological and biochemical parameters are observed; however, melatonin administration in radiotherapy plus melatonin groups improved these parameters (p <.05). In addition, there was no statistically significant difference between FF and FFF beams (p > .05). CONCLUSIONS The effect of low- and high-dose beams on the rat larynx and serum samples were investigated histopathologically and biochemically for the first time. We observed that melatonin supplemented before FF and FFF radiotherapy protected early period radiotherapy-induced laryngeal mucosal damage. Since the radiobiological results of FF and FFF beams are similar, FFF beams can be safely applied in laryngeal irradiation. However, more experimental rat and clinical studies are needed to clarify the radiobiological uncertainy concerning dose rate on cancerous and healthy tissue.
Collapse
Affiliation(s)
- Serhat Aras
- Medical Imaging Techniques Programme, University of Health Sciences Turkey, Istanbul, Turkey
| | - İhsan Oğuz Tanzer
- Biomedical Technology Programme, University of Health Sciences Turkey, Istanbul, Turkey.,Department of Neuroscience and Biomedical Engineering, Aalto University, Finland
| | - Neslihan Sayir
- Pathology Laboratory Techniques Programme, University of Health Sciences Turkey, Istanbul, Turkey
| | - Mevlüt Sait Keleş
- Department of Medical Biochemistry, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Fatma Betül Özgeriş
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Ataturk University, Erzurum, Turkey
| |
Collapse
|
40
|
Lenarczyk M, Laiakis EC, Mattson DL, Johnson BD, Kronenberg A, North PE, Komorowski R, Mäder M, Baker JE. Irradiation of the kidneys causes pathologic remodeling in the nontargeted heart: A role for the immune system. FASEB Bioadv 2020; 2:705-719. [PMID: 33336158 PMCID: PMC7734425 DOI: 10.1096/fba.2020-00071] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 08/26/2020] [Indexed: 12/17/2022] Open
Abstract
Cardiac disease is a frequent and significant adverse event associated with radiotherapy for cancer. Identifying the underlying mechanism responsible for radiation injury to the heart will allow interventions to be developed. In the present study, we tested if local kidney irradiation results in remodeling of the shielded, nontargeted heart. One kidney, two kidneys, or the total body of male WAG and Dahl SS rats were irradiated with 10 Gy of X-rays. Local kidney irradiation resulted in systemic hypertension, increased BUN, infiltration of T lymphocytes, natural killer cells, and macrophages into the renal cortex and medulla, and renal fibrosis. Local irradiation of kidneys in WAG rats resulted in remodeling in the nontargeted heart after 120 days, manifested by perivascular fibrosis and increased interventricular septal thickness, but was not seen in Dahl SS rats due to a high baseline level of fibrosis in the sham-irradiated animals. Genetic depletion of T cells mitigated the nephropathy after local kidney irradiation, indicating a role for the immune system in mediating this outcome. Local kidney irradiation resulted in a cascade of pro-inflammatory cytokines and low-molecular weight metabolites into the circulation associated with transmission of signals resulting in pathologic remodeling in the nontargeted heart. A new model is proposed whereby radiation-induced cardiac remodeling in susceptible animals is indirect, with lower hemi body organs such as the kidney exporting factors into the circulation that cause remodeling outside of the irradiated field in the shielded, nontargeted heart. This nontargeted effect appears to be mediated, in part, by the immune system.
Collapse
Affiliation(s)
| | | | | | | | - Amy Kronenberg
- Lawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | | | | | | | | |
Collapse
|
41
|
Turchan WT, Gutiontov SI, Spiotto MT, Liauw SL. Prostate Cancer Radiotherapy: Increased Biochemical Control and Late Toxicity in Men With Medication Allergies. JNCI Cancer Spectr 2020; 4:pkaa081. [PMID: 33409456 PMCID: PMC7771007 DOI: 10.1093/jncics/pkaa081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 11/13/2022] Open
Abstract
Abstract
Background
Given similarities in the mediators of medication allergy (MA) and tissue response to radiotherapy, we assessed whether outcomes following prostate radiotherapy differ in patients with MAs.
Methods
A total 587 men with known MA history and nonmetastatic prostate cancer underwent radiotherapy from 1989 to 2006. Clinicopathologic and treatment variables were analyzed for association with freedom from biochemical failure (FFBF) and late treatment–related, physician-defined Radiation Therapy Oncology Group gastrointestinal (GI) and genitourinary (GU) toxicity. Covariates identified on univariate analysis for toxicity and disease control were examined on multivariable analysis. All statistical tests were 2-sided, and a P less than .05 was considered statistically significant.
Results
A total of 155 of 587 men (26.4%) had 1 or more MAs, most commonly to penicillin (n = 71), sulfa (n = 35), and aspirin or nonsteroidal antiinflammatory drugs (n = 28). On univariate analysis, men with MAs had superior 10-y FFBF (71.5% vs 63.5%, P = .02) and higher incidence of late GI grade 2 or higher (G2+; 20.6% vs 13.2%, P = .04) and grade 3 or higher (G3+; 7.5% vs 3.9%, P = .08) as well as late GU G2+ (42.5% vs 33.2%, P = .04) and G3+ (7.5% vs 3.0%, P = .02) toxicity than men without MAs. On multivariable analysis, MA history remained a statistically significant predictor of FFBF (hazard ratio [HR] = 0.64, 95% confidence interval [CI] = 0.43 to 0.93, P = .02), late G2+ GI (HR = 1.76, 95% CI = 1.06 to 2.90, P=.03), and G3+ GU (HR = 2.69, 95% CI = 1.16 to 6.27, P = .02) toxicity after controlling for corresponding covariates in each model.
Conclusions
Men with MAs had improved FFBF and increased treatment-related toxicity following radiotherapy for prostate cancer. MA history could be a relevant consideration in the management of men with localized prostate cancer.
Collapse
Affiliation(s)
- William Tyler Turchan
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - Stanley I Gutiontov
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| | - Michael T Spiotto
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Stanley L Liauw
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
42
|
Anzic M, Marinko T. Effect of Adjuvant Hormonal Therapy on the Development of Pulmonary Fibrosis after Postoperative Radiotherapy for Breast Cancer. J Breast Cancer 2020; 23:449-459. [PMID: 33154822 PMCID: PMC7604379 DOI: 10.4048/jbc.2020.23.e48] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 08/19/2020] [Indexed: 12/15/2022] Open
Abstract
Breast cancer is the most common malignancy among women. Therefore, it is of paramount importance to study the adverse effects of oncological treatment of breast cancer, with one of adverse effects being pulmonary fibrosis (PF). PF is an irreversible condition and can significantly reduce the quality of life. Following lumpectomy, radiotherapy is the standard adjuvant treatment for breast cancer. Additionally, hormone receptor-positive breast cancers are treated with adjuvant hormonal therapy. While radiotherapy is one of the known causes of PF, the effect of hormone therapy on its development is not well-defined. Some studies have shown that the concomitant administration of endocrine therapy, primarily tamoxifen, and irradiation may potentiate the development of PF. However, guidelines regarding the timing of hormone therapy administration with respect to adjuvant radiotherapy are not clearly defined. This review aims to provide a comprehensive overview of the available information regarding the effect of hormone therapy and its timing of administration with respect to adjuvant radiotherapy on the incidence of PF.
Collapse
Affiliation(s)
- Mitja Anzic
- Division of Radiotherapy, Institute of Oncology, Ljubljana, Slovenia
| | - Tanja Marinko
- Division of Radiotherapy, Institute of Oncology, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
43
|
Evaluation of cytokine expression and circulating immune cell subsets as potential parameters of acute radiation toxicity in prostate cancer patients. Sci Rep 2020; 10:19002. [PMID: 33149212 PMCID: PMC7643057 DOI: 10.1038/s41598-020-75812-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 10/09/2020] [Indexed: 01/02/2023] Open
Abstract
One of the challenges of radiation oncology in the era of personalized medicine is identification of biomarkers associated with individual radiosensitivity. The aim of research was to evaluate the possible clinical value of the associations between clinical, physical, and biological factors, and risk for development of acute radiotoxicity in patients with prostate cancer. The study involved forty four patients treated with three-dimensional conformal radiotherapy. The concentrations of IL-1β, IL-2, IL-6, IFN-γ and TGF-β1 were assessed before radiotherapy, after 5th, 15th and 25th radiotherapy fractions, at the end, and 1 month after the end of radiotherapy. Cytokine gene expression was determined in peripheral blood mononuclear cells. The univariate analysis of circulating cytokine levels during radiotherapy showed that increased serum concentrations of IL-6 were significantly associated with higher grade of acute genitourinary toxicity. The multivariate analysis demonstrated that increased level of IL-6 during the radiotherapy was significantly associated with higher grade of acute genitourinary toxicity across treatment. TGF-β expression levels significantly decreased during course of radiotherapy. Research indicates that changes in circulating cytokine levels might be important parameter of radiotoxicity in patients with prostate cancer. These findings suggest that future studies based on multi-parameter examination are necessary for prediction of individual radiosensitivity.
Collapse
|
44
|
Nakamura N. A hypothesis: radiation carcinogenesis may result from tissue injuries and subsequent recovery processes which can act as tumor promoters and lead to an earlier onset of cancer. Br J Radiol 2020; 93:20190843. [PMID: 31860335 PMCID: PMC8519633 DOI: 10.1259/bjr.20190843] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cancer risks from radiation can be observed as an increase in mortality when compared to a control group. However, it is unknown if this increased risk results from the induction of cancer or from an earlier onset of cancer. In mouse studies, it has been repeatedly shown that after an irradiation, the survival curve is shifted toward lower ages, but remains parallel to the control curve, and the extent of the shift in time to lower ages is dose-dependent. This shift is not satisfactorily explained by the induction model which assumes that cancers in the exposed group consist of spontaneous and induced events. Consequently, it seems that this shift could be interpreted to mean that all animals in the exposed group had suffered from life shortening. Under this scenario, however, it turns out that the radiation effects can no longer be interpreted as the result of oncogenic mutations, because these effects would have to involve all tumors, and the effectiveness of radiation changes with the dose. This leads to the speculation that radiation exposures induce a broad range of tissue injuries, and that these injuries are subsequently subjected to longlasting systemic recovery processes which act as promoters for tumor cells. In other words, potential cancer stem cells which were located in the irradiated field can escape oncogenic damage but undergo stimulation later in life toward the development of malignancy from radiation-induced activated microenvironment. This is an unusual form of the non-targeted or bystander effects of radiation. It is worth noting that this model suggests that there could be a path or paths which could be used to intervene in the process of post-exposure carcinogenesis, and that cancer risks at low doses could be described as days or weeks of life lost.
Collapse
Affiliation(s)
- Nori Nakamura
- Dept. of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima city, Japan
| |
Collapse
|
45
|
PM014 attenuates radiation-induced pulmonary fibrosis via regulating NF-kB and TGF-b1/NOX4 pathways. Sci Rep 2020; 10:16112. [PMID: 32999298 PMCID: PMC7527517 DOI: 10.1038/s41598-020-72629-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 09/04/2020] [Indexed: 12/31/2022] Open
Abstract
Radiation therapy is the mainstay in the treatment of lung cancer, and lung fibrosis is a radiotherapy-related major side effect that can seriously reduce patient’s quality of life. Nevertheless, effective strategies for protecting against radiation therapy-induced fibrosis have not been developed. Hence, we investigated the radioprotective effects and the underlying mechanism of the standardized herbal extract PM014 on radiation-induced lung fibrosis. Ablative radiation dose of 75 Gy was focally delivered to the left lung of mice. We evaluated the effects of PM014 on radiation-induced lung fibrosis in vivo and in an in vitro model. Lung volume and functional changes were evaluated using the micro-CT and flexiVent system. Fibrosis-related molecules were evaluated by immunohistochemistry, western blot, and real-time PCR. A orthotopic lung tumour mouse model was established using LLC1 cells. Irradiated mice treated with PM014 showed a significant improvement in collagen deposition, normal lung volume, and functional lung parameters, and these therapeutic effects were better than those of amifostine. PM104 attenuated radiation-induced increases in NF-κB activity and inhibited radiation-induced p65 translocation, ROS production, DNA damage, and epithelial-mesenchymal transition. PM104 effectively alleviated fibrosis in an irradiated orthotopic mouse lung tumour model while not attenuating the efficacy of the radiation therapy by reduction of the tumour. Standardized herbal extract PM014 may be a potential therapeutic agent that is able to increase the efficacy of radiotherapy by alleviating radiation-induced lung fibrosis.
Collapse
|
46
|
Nielsen S, Bassler N, Grzanka L, Swakon J, Olko P, Horsman MR, Sørensen BS. Proton scanning and X-ray beam irradiation induce distinct regulation of inflammatory cytokines in a preclinical mouse model. Int J Radiat Biol 2020; 96:1238-1244. [PMID: 32780616 DOI: 10.1080/09553002.2020.1807644] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE Conventional X-ray radiotherapy induces a pro-inflammatory response mediated by altered expression of inflammation-regulating cytokines. Proton scanning and X-ray irradiation produce distinct changes to cytokine gene expression in vitro suggesting that proton beam therapy may induce an inflammatory response dissimilar to that of X-ray radiation. The purpose of the present study was to determine whether proton scanning beam radiation and conventional X-ray photon radiation would induce differential regulation of circulating cytokines in vivo. MATERIALS AND METHODS Female CDF1 mice were irradiated locally at the right hind leg using proton pencil beam scanning or X-ray photons. Blood samples were obtained from two separate mice groups. Samples from one group were drawn by retro-orbital puncture 16 months post irradiation, while samples from the other group were drawn 5 and 30 days post irradiation. Concentration of the cytokines IL-6, IL-1β, IL-10, IL-17A, IFN-γ, and TNFα was measured in plasma using bead-based immunoassays. RESULTS The cytokines IL-6, IL-1β, IL-10, IFN-γ, and TNFα were expressed at lower levels in plasma samples from proton-irradiated mice compared with X-ray-irradiated mice 16 months post irradiation. The same cytokines were downregulated in proton-irradiated mice 5 days post irradiation when compared to controls, while at day 30 expression had increased to the same level or higher. X-ray radiation did not markedly change expression levels at days 5 and 30. CONCLUSIONS The inflammatory response to proton and X-ray irradiation seem to be distinct as the principal pro-inflammatory cytokines are differentially regulated short- and long-term following irradiation. Both the development of normal tissue damage and efficacy of immunotherapy could be influenced by an altered inflammatory response to irradiation.
Collapse
Affiliation(s)
- Steffen Nielsen
- Experimental Clinical Oncology, Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Niels Bassler
- Medical Radiation Physics, Department of Physics, Stockholm University, Stockholm, Sweden
| | - Leszek Grzanka
- Proton Radiotherapy Group, Institute of Nuclear Physics, Polish Academy of Sciences, Krakow, Poland
| | - Jan Swakon
- Proton Radiotherapy Group, Institute of Nuclear Physics, Polish Academy of Sciences, Krakow, Poland
| | - Pawel Olko
- Proton Radiotherapy Group, Institute of Nuclear Physics, Polish Academy of Sciences, Krakow, Poland
| | - Michael R Horsman
- Experimental Clinical Oncology, Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Brita Singers Sørensen
- Experimental Clinical Oncology, Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
47
|
Käsmann L, Dietrich A, Staab-Weijnitz CA, Manapov F, Behr J, Rimner A, Jeremic B, Senan S, De Ruysscher D, Lauber K, Belka C. Radiation-induced lung toxicity - cellular and molecular mechanisms of pathogenesis, management, and literature review. Radiat Oncol 2020; 15:214. [PMID: 32912295 PMCID: PMC7488099 DOI: 10.1186/s13014-020-01654-9] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/20/2020] [Indexed: 12/17/2022] Open
Abstract
Lung, breast, and esophageal cancer represent three common malignancies with high incidence and mortality worldwide. The management of these tumors critically relies on radiotherapy as a major part of multi-modality care, and treatment-related toxicities, such as radiation-induced pneumonitis and/or lung fibrosis, are important dose limiting factors with direct impact on patient outcomes and quality of life. In this review, we summarize the current understanding of radiation-induced pneumonitis and pulmonary fibrosis, present predictive factors as well as recent diagnostic and therapeutic advances. Novel candidates for molecularly targeted approaches to prevent and/or treat radiation-induced pneumonitis and pulmonary fibrosis are discussed.
Collapse
Affiliation(s)
- Lukas Käsmann
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany.
- German Center for Lung Research (DZL), partner site Munich, Munich, Germany.
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany.
| | - Alexander Dietrich
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), Medical Faculty, LMU-Munich, Munich, Germany
| | - Claudia A Staab-Weijnitz
- German Center for Lung Research (DZL), partner site Munich, Munich, Germany
- Institute of Lung Biology and Disease, Helmholtz Zentrum München, Munich, Germany
| | - Farkhad Manapov
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
- German Center for Lung Research (DZL), partner site Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
| | - Jürgen Behr
- German Center for Lung Research (DZL), partner site Munich, Munich, Germany
- Department of Internal Medicine V, LMU Munich, Munich, Germany
| | - Andreas Rimner
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, USA
| | | | - Suresh Senan
- Department of Radiation Oncology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Dirk De Ruysscher
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Kirsten Lauber
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistrasse 15, 81377, Munich, Germany
- German Center for Lung Research (DZL), partner site Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
| |
Collapse
|
48
|
Patel P, Malipatlolla DK, Devarakonda S, Bull C, Rascón A, Nyman M, Stringer A, Tremaroli V, Steineck G, Sjöberg F. Dietary Oat Bran Reduces Systemic Inflammation in Mice Subjected to Pelvic Irradiation. Nutrients 2020; 12:nu12082172. [PMID: 32707913 PMCID: PMC7468988 DOI: 10.3390/nu12082172] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/10/2020] [Accepted: 07/18/2020] [Indexed: 12/14/2022] Open
Abstract
Patients undergoing radiotherapy to treat pelvic-organ cancer are commonly advised to follow a restricted fiber diet. However, reducing dietary fiber may promote gastrointestinal inflammation, eventually leading to deteriorated intestinal health. The goal of this study was to evaluate the influence of dietary fiber on radiation-induced inflammation. C57BL/6J male mice were fed a High-oat bran diet (15% fiber) or a No-fiber diet (0% fiber) and were either irradiated (32 Gy delivered in four fractions) to the colorectal region or only sedated (controls). The dietary intervention started at 2 weeks before irradiation and lasted for 1, 6, and 18 weeks after irradiation, at which time points mice were sacrificed and their serum samples were assayed for 23 cytokines and chemokines. Our analyses show that irradiation increased the serum cytokine levels at all the time points analyzed. The No-fiber irradiated mice had significantly higher levels of pro-inflammatory cytokines than the High-oat irradiated mice at all time points. The results indicate that a fiber-rich oat bran diet reduces the intensity of radiation-induced inflammation, both at an early and late stage. Based on the results, it seems that the advice to follow a low-fiber diet during radiotherapy may increase the risk of decreased intestinal health in cancer survivors.
Collapse
Affiliation(s)
- Piyush Patel
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden; (D.K.M.); (S.D.); (C.B.); (G.S.); (F.S.)
- Department of Infectious Diseases, Institute of Biomedicine, the Sahlgrenska Academy, University of Gothenburg, 41346 Gothenburg, Sweden
- Correspondence:
| | - Dilip Kumar Malipatlolla
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden; (D.K.M.); (S.D.); (C.B.); (G.S.); (F.S.)
| | - Sravani Devarakonda
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden; (D.K.M.); (S.D.); (C.B.); (G.S.); (F.S.)
| | - Cecilia Bull
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden; (D.K.M.); (S.D.); (C.B.); (G.S.); (F.S.)
| | - Ana Rascón
- Department of Food Technology, Engineering and Nutrition, Lund University, 22100 Lund, Sweden; (A.R.); (M.N.)
| | - Margareta Nyman
- Department of Food Technology, Engineering and Nutrition, Lund University, 22100 Lund, Sweden; (A.R.); (M.N.)
| | - Andrea Stringer
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide SA 5001, Australia;
| | - Valentina Tremaroli
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, the Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden;
| | - Gunnar Steineck
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden; (D.K.M.); (S.D.); (C.B.); (G.S.); (F.S.)
| | - Fei Sjöberg
- Division of Clinical Cancer Epidemiology, Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden; (D.K.M.); (S.D.); (C.B.); (G.S.); (F.S.)
- Department of Infectious Diseases, Institute of Biomedicine, the Sahlgrenska Academy, University of Gothenburg, 41346 Gothenburg, Sweden
| |
Collapse
|
49
|
Barazzuol L, Coppes RP, van Luijk P. Prevention and treatment of radiotherapy-induced side effects. Mol Oncol 2020; 14:1538-1554. [PMID: 32521079 PMCID: PMC7332214 DOI: 10.1002/1878-0261.12750] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 05/29/2020] [Accepted: 06/02/2020] [Indexed: 01/10/2023] Open
Abstract
Radiotherapy remains a mainstay of cancer treatment, being used in roughly 50% of patients. The precision with which the radiation dose can be delivered is rapidly improving. This precision allows the more accurate targeting of radiation dose to the tumor and reduces the amount of surrounding normal tissue exposed. Although this often reduces the unwanted side effects of radiotherapy, we still need to further improve patients' quality of life and to escalate radiation doses to tumors when necessary. High-precision radiotherapy forces one to choose which organ or functional organ substructures should be spared. To be able to make such choices, we urgently need to better understand the molecular and physiological mechanisms of normal tissue responses to radiotherapy. Currently, oversimplified approaches using constraints on mean doses, and irradiated volumes of normal tissues are used to plan treatments with minimized risk of radiation side effects. In this review, we discuss the responses of three different normal tissues to radiotherapy: the salivary glands, cardiopulmonary system, and brain. We show that although they may share very similar local cellular processes, they respond very differently through organ-specific, nonlocal mechanisms. We also discuss how a better knowledge of these mechanisms can be used to treat or to prevent the effects of radiotherapy on normal tissue and to optimize radiotherapy delivery.
Collapse
Affiliation(s)
- Lara Barazzuol
- Department of Biomedical Sciences of Cells and SystemsUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
- Department of Radiation OncologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Rob P. Coppes
- Department of Biomedical Sciences of Cells and SystemsUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
- Department of Radiation OncologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Peter van Luijk
- Department of Biomedical Sciences of Cells and SystemsUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
- Department of Radiation OncologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| |
Collapse
|
50
|
Jin H, Yoo Y, Kim Y, Kim Y, Cho J, Lee YS. Radiation-Induced Lung Fibrosis: Preclinical Animal Models and Therapeutic Strategies. Cancers (Basel) 2020; 12:cancers12061561. [PMID: 32545674 PMCID: PMC7352529 DOI: 10.3390/cancers12061561] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/02/2020] [Accepted: 06/10/2020] [Indexed: 01/27/2023] Open
Abstract
Radiation-induced lung injury (RILI), including acute radiation pneumonitis and chronic radiation-induced lung fibrosis, is the most common side effect of radiation therapy. RILI is a complicated process that causes the accumulation, proliferation, and differentiation of fibroblasts and, finally, results in excessive extracellular matrix deposition. Currently, there are no approved treatment options for patients with radiation-induced pulmonary fibrosis (RIPF) partly due to the absence of effective targets. Current research advances include the development of small animal models reflecting modern radiotherapy, an understanding of the molecular basis of RIPF, and the identification of candidate drugs for prevention and treatment. Insights provided by this research have resulted in increased interest in disease progression and prognosis, the development of novel anti-fibrotic agents, and a more targeted approach to the treatment of RIPF.
Collapse
Affiliation(s)
- Hee Jin
- Graduate School of Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University, Seoul 03760, Korea; (H.J.); (Y.Y.); (Y.K.); (Y.K.)
| | - Youngjo Yoo
- Graduate School of Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University, Seoul 03760, Korea; (H.J.); (Y.Y.); (Y.K.); (Y.K.)
| | - Younghwa Kim
- Graduate School of Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University, Seoul 03760, Korea; (H.J.); (Y.Y.); (Y.K.); (Y.K.)
| | - Yeijin Kim
- Graduate School of Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University, Seoul 03760, Korea; (H.J.); (Y.Y.); (Y.K.); (Y.K.)
| | - Jaeho Cho
- Department of Radiation Oncology, Yonsei University Health System, Seoul 03722, Korea
- Correspondence: (J.C.); (Y.-S.L.); Tel.: +82-2-2228-8113 (J.C.); +82-2-3277-3022 (Y.-S.L.); Fax: +82-2-3277-3051 (Y.-S.L.)
| | - Yun-Sil Lee
- Graduate School of Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University, Seoul 03760, Korea; (H.J.); (Y.Y.); (Y.K.); (Y.K.)
- Correspondence: (J.C.); (Y.-S.L.); Tel.: +82-2-2228-8113 (J.C.); +82-2-3277-3022 (Y.-S.L.); Fax: +82-2-3277-3051 (Y.-S.L.)
| |
Collapse
|