1
|
Nagendra S, Gutte AA, Gaikwad AM. Amaurosis Fugax Due to Radiation-Induced Carotid Stenosis Treated by Carotid Artery Stenting. Neurol India 2025:02223311-990000000-00086. [PMID: 40238660 DOI: 10.4103/neuroindia.ni_472_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/17/2020] [Indexed: 04/18/2025]
Affiliation(s)
- Shashank Nagendra
- Department of Neurology, Grant Medical College and JJ Hospital, Mumbai, Maharashtra, India
| | - Avinash A Gutte
- Department of Interventional Radiology, Grant Medical College and JJ Hospital, Mumbai, Maharashtra, India
| | - Abhijit M Gaikwad
- Department of Neurology, Grant Medical College and JJ Hospital, Mumbai, Maharashtra, India
| |
Collapse
|
2
|
Zaghlol R, Pedersen L, Qamer S, Yoo SGK, Ladin DA, Parvathaneni A, Bergom C, Mitchell JD. Cardiac Complications of Radiation Therapy. Cardiol Clin 2025; 43:129-149. [PMID: 39551554 DOI: 10.1016/j.ccl.2024.09.007] [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] [Indexed: 11/19/2024]
Abstract
Radiation therapy is a critical component in managing many malignancies by improving local control and survival. The benefits of radiation may come at the expense of unintended radiation injury to the surrounding normal tissues, with the heart being one of the most affected organs in thoracic radiation treatments. As cancer survivors live longer, radiation-induced cardiotoxicity (RICT) is now increasingly recognized. In this review, we highlight the spectrum and pathophysiology of RICT. We summarize contemporary recommendations for risk stratification, screening, prevention, and management of RICT. We briefly highlight novel applications for radiation to treat some cardiac conditions such as resistant arrhythmias.
Collapse
Affiliation(s)
- Raja Zaghlol
- Division of Cardiovascular Disease, Cardio-oncology Section, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8086, St Louis, MO 63110, USA
| | - Lauren Pedersen
- Department of Radiation Oncology, Washington University School of Medicine, Alvin J. Siteman Cancer Center, Washington University in St. Louis, 4511 Forest Park Avenue, Suite 3106A, St. Louis, MO 63108, USA
| | - Syed Qamer
- Division of Cardiovascular Disease, Cardio-oncology Section, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8086, St Louis, MO 63110, USA
| | - Sang Gune K Yoo
- Division of Cardiovascular Disease, Cardio-oncology Section, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8086, St Louis, MO 63110, USA
| | - Daniel A Ladin
- John T. Milliken Department of Medicine, Washington University in St. Louis, Saint Louis, MO 63110, USA
| | - Adeesh Parvathaneni
- Center for Cardiovascular Research, Schilling Lab, Washington University School of Medicine in St. Louis, St Louis, MO 63110, USA
| | - Carmen Bergom
- Department of Radiation Oncology, Washington University School of Medicine, Alvin J. Siteman Cancer Center, Washington University in St. Louis, 4511 Forest Park Avenue, Suite 3106A, St. Louis, MO 63108, USA
| | - Joshua D Mitchell
- Division of Cardiovascular Disease, Cardio-oncology Section, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8086, St Louis, MO 63110, USA.
| |
Collapse
|
3
|
Giacinto O, Pelliccia F, Minati A, De Crescenzo F, Garo ML, Chello M, Lusini M. Cosmic Radiations and the Cardiovascular System: A Narrative Review. Cardiol Rev 2024; 32:433-439. [PMID: 36728769 DOI: 10.1097/crd.0000000000000521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
In recent times, space flights receive continued interest. Humankind's next two goals are to return to the Moon and, a few years later, to land on the surface of Mars. Although technology will improve enough to enable long voyages, there are still some unresolved questions about the effects of the space environment on human health, including the effects of such long voyages on organs. Specifically, there is no information on the effects of radiation in space on the human cardiovascular system. To better understand the adaptation of the cardiovascular system to radiation exposure, the physical properties of radiation and the cellular and molecular mechanisms underlying tissue changes are essential. To this end, this article aims to provide an overview of the effects of radiation on the cardiovascular system by analyzing the physical properties of radiation and their relationship to cellular and molecular mechanisms and potential changes. Each type of radiation triggers different responses in the cardiovascular system. Radiation plays a relevant role in altering endothelial function and arterial wall stiffness by inducing vascular changes that accelerate atherosclerosis and affect endothelial adhesiveness. Clinical studies have shown that vascular changes due to radiation depend on the delayed manifestations of early radiation damage. To reduce the effects of radiation in space, some pharmacological treatments that seem to be able to counteract oxidative stress during flight are being used. At the same time, new shielding systems that can reduce or eliminate radiation exposure must be developed. Future studies should aim to replicate flights in the deep space environment to study in more detail the harmful effects of radiation on the whole cardiovascular system.
Collapse
Affiliation(s)
- Omar Giacinto
- From the Università Campus Bio-medico di Roma, UOC di Cardiochirurgia, Rome, Italy
| | | | | | | | - Maria Luisa Garo
- From the Università Campus Bio-medico di Roma, UOC di Cardiochirurgia, Rome, Italy
| | - Massimo Chello
- From the Università Campus Bio-medico di Roma, UOC di Cardiochirurgia, Rome, Italy
| | - Mario Lusini
- From the Università Campus Bio-medico di Roma, UOC di Cardiochirurgia, Rome, Italy
| |
Collapse
|
4
|
Kiang JG, Cannon G, Singh VK. An Overview of Radiation Countermeasure Development in Radiation Research from 1954 to 2024. Radiat Res 2024; 202:420-431. [PMID: 38964743 PMCID: PMC11385179 DOI: 10.1667/rade-24-00036.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 03/21/2024] [Indexed: 07/06/2024]
Abstract
Preparation for medical responses to major radiation accidents, further driven by increases in the threat of nuclear warfare, has led to a pressing need to understand the underlying mechanisms of radiation injury (RI) alone or in combination with other trauma (combined injury, CI). The identification of these mechanisms suggests molecules and signaling pathways that can be targeted to develop radiation medical countermeasures. Thus far, the United States Food and Drug Administration (U.S. FDA) has approved seven countermeasures to mitigate hematopoietic acute radiation syndrome (H-ARS), but no drugs are available for prophylaxis and no agents have been approved to combat the other sub-syndromes of ARS, let alone delayed effects of acute radiation exposure or the effects of combined injury. From its inception, Radiation Research has significantly contributed to the understanding of the underlying mechanisms of radiation injury and combined injury, and to the development of radiation medical countermeasures for these indications through the publication of peer-reviewed research and review articles.
Collapse
Affiliation(s)
- Juliann G Kiang
- Scientific Research Department, Armed Forces Radiobiology Research Institute
- Department of Pharmacology and Molecular Therapeutics, School of Medicine
- Department of Medicine, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Georgetta Cannon
- Scientific Research Department, Armed Forces Radiobiology Research Institute
| | - Vijay K Singh
- Scientific Research Department, Armed Forces Radiobiology Research Institute
- Department of Pharmacology and Molecular Therapeutics, School of Medicine
| |
Collapse
|
5
|
Chen Y, Xu Y, Pan D, Li H, Cai J, Li Y, Shen Q, Tang Y. Progression rate of radiation-induced carotid stenosis in head and neck cancer survivors after statin treatment: a retrospective cohort study. J Neurol 2024; 271:2573-2581. [PMID: 38332351 DOI: 10.1007/s00415-024-12197-4] [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: 10/16/2023] [Revised: 01/03/2024] [Accepted: 01/11/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND AND AIMS Whether statin treatment is effective in retarding the progression of radiation-induced carotid stenosis (RICS) in head and neck cancer (HNC) survivors has not been well studied. The purpose of this study was to assess the association of statin treatment with RICS progression rate in HNC survivors after radiotherapy. METHODS We conducted a retrospective cohort study at Sun Yat-sen Memorial Hospital, Sun Yat-sen University in Guangzhou, China. Between January 2010 and December 2021, we screened HNC survivors whose carotid ultrasound scans had shown stenosis of the common and/or internal carotid arteries. The primary outcome was the RICS progression rate. We compared eligible patients treated with statins with those who did not in multivariable Cox regression models. RESULTS A total of 200 patients were included in this study, of whom 108 received statin treatment and 92 did not. Over a mean follow-up time of 1.5 years, 56 (28.0%) patients showed RICS progression, 24 (42.9%) and 32 (57.1%) in the statin and control groups, respectively. The statin group showed less RICS progression than the control group (adjusted-HR 0.49, 95% CI 0.30-0.80, P = 0.005). In the subgroup analysis, there was no significant interaction in the effect of statins on lowering RICS progression rate in the subgroups stratified by baseline low-density lipoprotein cholesterol (LDL-C) levels (P for interaction = 0.53) or baseline degrees of stenosis (P for interaction = 0.50). CONCLUSIONS Statin treatment was associated with a lower risk of RICS progression in patients with HNC after radiotherapy, regardless of baseline LDL-C level and baseline stenosis degrees.
Collapse
Affiliation(s)
- Yanting Chen
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yongteng Xu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Dong Pan
- Department of Neurology, The Eighth Affiliated Hospital, SunYat-Sen University, Shenzhen, 528406, China
| | - Honghong Li
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jinhua Cai
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yi Li
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Qingyu Shen
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Yamei Tang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510120, China.
| |
Collapse
|
6
|
Yoo J, Jeon J, Baik M, Kim J. Risk of thromboembolism according to statin treatment in patients with cancer: A nationwide nested case-control study. Thromb Res 2024; 235:32-40. [PMID: 38295599 DOI: 10.1016/j.thromres.2024.01.016] [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: 10/02/2023] [Revised: 01/12/2024] [Accepted: 01/19/2024] [Indexed: 02/02/2024]
Abstract
BACKGROUND Thromboembolic events exhibit increased prevalence in patients with cancer and can negatively affect prognoses. We investigated whether statin treatment would reduce thromboembolic risk in patients with cancer. METHODS We conducted a nested case-control study using a Korean nationwide health claims database. The study included patients newly diagnosed with cancer without a prior history of cardiovascular disease between 2014 and 2016. Cases who developed arterial thromboembolism (ATE) or venous thromboembolism (VTE) after cancer diagnosis and three individually matched controls were selected. Conditional logistic regression was used to assess the association between thromboembolic risk and statin therapy after cancer diagnosis. RESULTS Among 455,805 newly diagnosed patients with cancer followed for a mean of 4.3 ± 2.0 years, 22,249 patients developed thromboembolic events (ATE: 6341, VTE: 15,908), resulting in an incidence rate of 1133 per 100,000 person-years. The nested case-control study included 21,289 cases with thromboembolic events and 63,867 controls. Statin use was less frequent in the case group (18.0 % vs. 23.7 %). Statin treatment was associated with a lower risk of thromboembolic events (adjusted odds ratio [OR] 0.70; 95 % confidence interval [CI] 0.67-0.73). This association was observed for both ATE (adjusted OR 0.68; 95 % CI 0.63-0.74) and VTE (adjusted OR 0.71; 95 % CI 0.67-0.75). Longer statin use and better adherence were also associated with lower risk for thromboembolic events. Statin treatment was significantly associated with fewer thromboembolic events in most cancer types. CONCLUSIONS Statin use was associated with lower risk for thromboembolic events in patients newly diagnosed with cancer.
Collapse
Affiliation(s)
- Joonsang Yoo
- Department of Neurology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Republic of Korea
| | - Jimin Jeon
- Department of Neurology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Republic of Korea
| | - Minyoul Baik
- Department of Neurology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Republic of Korea
| | - Jinkwon Kim
- Department of Neurology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Republic of Korea; Institute for Innovation in Digital Healthcare, Yonsei University Health System, Seoul, Republic of Korea.
| |
Collapse
|
7
|
Guo X, Osouli S, Shahripour RB. Review of Cerebral Radiotherapy-Induced Vasculopathy in Pediatric and Adult Patients. Adv Biol (Weinh) 2023; 7:e2300179. [PMID: 37401794 DOI: 10.1002/adbi.202300179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/04/2023] [Indexed: 07/05/2023]
Abstract
Radiation therapy (RT) causes radiation-induced vasculopathy, which requires clinicians to identify and manage this side effect in pediatric and adult patients. This article reviews previous findings about the pathophysiology of RT-induced vascular injury, including endothelial cell injury, oxidative stress, inflammatory cytokines, angiogenic pathways, and remodeling. The vasculopathy is categorized into ischemic vasculopathy, hemorrhagic vasculopathy, carotid artery injury, and other malformations (cavernous malformations and aneurysms) in populations of pediatric and adult patients separately. The prevention and management of this RT-induced side effect are also discussed. The article summarizes the distribution and risk factors of different types of RT-induced vasculopathy. This will help clinicians identify high-risk patients with corresponding vasculopathy subtypes to deduce prevention and treatment strategies accordingly.
Collapse
Affiliation(s)
- Xiaofan Guo
- Department of Neurology, Loma Linda University, Loma Linda, CA, 92354, USA
| | - Sima Osouli
- Department of Neurology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1516745811, Iran
| | - Reza Bavarsad Shahripour
- Department of Neurology, Loma Linda University, Loma Linda, CA, 92354, USA
- Comprehensive Stroke Center, Department of Neurology, University of California San Diego, San Diego, CA, 92103, USA
| |
Collapse
|
8
|
Kim JM, Kim H, Oh SH, Jang WI, Lee SB, Park M, Kim S, Park S, Shim S, Jang H. Combined Administration of Pravastatin and Metformin Attenuates Acute Radiation-Induced Intestinal Injury in Mouse and Minipig Models. Int J Mol Sci 2022; 23:ijms232314827. [PMID: 36499155 PMCID: PMC9739896 DOI: 10.3390/ijms232314827] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 12/02/2022] Open
Abstract
Radiation-induced gastrointestinal (GI) damage is one of the critical factors that serve as basis for the lethality of nuclear accidents or terrorism. Further, there are no Food and Drug Administration-approved agents available to mitigate radiation-induced intestinal injury. Although pravastatin (PS) has been shown to exhibit anti-inflammatory and epithelial reconstructive effects following radiation exposure using mouse and minipig models, the treatment failed to improve the survival rate of high-dose irradiated intestinal injury. Moreover, we previously found that metformin (MF), a common drug used for treating type 2 diabetes mellitus, has a mitigating effect on radiation-induced enteropathy by promoting stem cell properties. In this study, we investigated whether the combined administration of PS and MF could achieve therapeutic effects on acute radiation-induced intestinal injury in mouse and minipig models. We found that the combined treatment markedly increased the survival rate and attenuated histological damage in a radiation-induced intestinal injury mouse model, in addition to epithelial barrier recovery, anti-inflammatory effects, and improved epithelial proliferation with stem cell properties. Furthermore, in minipig models, combined treatment with PS and MF ameliorates gross pathological damage in abdominal organs and attenuated radiation-induced intestinal histological damage. Therefore, the combination of PS and MF effectively alleviated radiation-induced intestinal injury in the mouse and minipig models. We believe that the combined use of PS and MF is a promising therapeutic approach for treating radiation-induced intestinal injury.
Collapse
Affiliation(s)
- Jung Moon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
- Department of Veterinary Surgery, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Hyewon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
| | - Su Hyun Oh
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
| | - Won Il Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
| | - Seung Bum Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
| | - Mineon Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
| | - Soyeon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
| | - Sunhoo Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
| | - Sehwan Shim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
- Correspondence: (S.S.); (H.J.); Tel.: +82-2-3399-5873 (S.S.); +82-2-970-1302 (H.J.)
| | - Hyosun Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Science, Seoul 01812, Republic of Korea
- Correspondence: (S.S.); (H.J.); Tel.: +82-2-3399-5873 (S.S.); +82-2-970-1302 (H.J.)
| |
Collapse
|
9
|
The Prediction of Cardiac Events Using Contemporary Risk Prediction Models after Radiation Therapy for Head and Neck Cancer. Cancers (Basel) 2022; 14:cancers14153651. [PMID: 35954315 PMCID: PMC9367378 DOI: 10.3390/cancers14153651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/01/2022] [Accepted: 07/15/2022] [Indexed: 02/04/2023] Open
Abstract
This study aims to evaluate the efficacy of the Pooled Cohort Equation (PCE), U.S. Preventative Services Task Force (USPSTF), and Framingham Risk Score (FRS) models in predicting ASCVD events among patients receiving radiation therapy (RT) for head and neck cancer (HNCA). From a large cohort of HNCA patients treated with RT, ASCVD events were adjudicated. Observed vs. predicted ASCVD events were compared. We compared rates by statin eligibility status. Regression models and survival analysis were used to identify the relationship between predicted risk and post-RT outcomes. Among the 723 identified patients, 274 (38%) were statin-eligible based on USPSTF criteria, 359 (49%) based on PCE, and 234 (32%) based on FRS. During follow-up, 17% developed an ASCVD, with an event rate of 27 per 1000 person-years, 68% higher than predicted (RR 1.68 (95% CI: 1.02, 2.12), p < 0.001). In multivariable regression, there was no difference in event rates by statin eligibility status (p > 0.05). Post-RT, the observed event rate was higher than the predicted ASCVD risk across all grades of predicted risk (p < 0.05) and the observed risk of an ASCVD event was high even among patients predicted to have a low risk of ASCVD. In conclusion, current ASCVD risk calculators significantly underestimate the risk for ASCVD among patients receiving RT for HNCA.
Collapse
|
10
|
Badescu MC, Badulescu OV, Scripcariu DV, Butnariu LI, Bararu-Bojan I, Popescu D, Ciocoiu M, Gorduza EV, Costache II, Rezus E, Rezus C. Myocardial Ischemia Related to Common Cancer Therapy-Prevention Insights. Life (Basel) 2022; 12:1034. [PMID: 35888122 PMCID: PMC9325217 DOI: 10.3390/life12071034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/08/2022] [Accepted: 07/10/2022] [Indexed: 12/18/2022] Open
Abstract
Modern antineoplastic therapy improves survival and quality of life in cancer patients, but its indisputable benefits are accompanied by multiple and major side effects, such as cardiovascular ones. Endothelial dysfunction, arterial spasm, intravascular thrombosis, and accelerated atherosclerosis affect the coronary arteries, leading to acute and chronic coronary syndromes that negatively interfere with the oncologic treatment. The cardiac toxicity of antineoplastic agents may be mitigated by using adequate prophylactic measures. In the absence of dedicated guidelines, our work provides the most comprehensive, systematized, structured, and up-to-date analyses of the available literature focusing on measures aiming to protect the coronary arteries from the toxicity of cancer therapy. Our work facilitates the implementation of these measures in daily practice. The ultimate goal is to offer clinicians the necessary data for a personalized therapeutic approach for cancer patients receiving evidence-based oncology treatments with potential cardiovascular toxicity.
Collapse
Affiliation(s)
- Minerva Codruta Badescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (M.C.B.); (D.P.); (I.I.C.); (C.R.)
- III Internal Medicine Clinic, “St. Spiridon” County Emergency Clinical Hospital, 1 Independence Boulevard, 700111 Iasi, Romania
| | - Oana Viola Badulescu
- Department of Pathophysiology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.B.-B.); (M.C.)
- Hematology Clinic, “St. Spiridon” County Emergency Clinical Hospital, 1 Independence Boulevard, 700111 Iasi, Romania
| | - Dragos Viorel Scripcariu
- Surgery Department, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
- 1st Surgical Oncology Unit, Regional Institute of Oncology, 2-4 General Henri Mathias Berthelot Street, 700483 Iasi, Romania
| | - Lăcrămioara Ionela Butnariu
- Department of Mother and Child Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Iris Bararu-Bojan
- Department of Pathophysiology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.B.-B.); (M.C.)
| | - Diana Popescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (M.C.B.); (D.P.); (I.I.C.); (C.R.)
| | - Manuela Ciocoiu
- Department of Pathophysiology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.B.-B.); (M.C.)
| | - Eusebiu Vlad Gorduza
- Department of Mother and Child Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Irina Iuliana Costache
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (M.C.B.); (D.P.); (I.I.C.); (C.R.)
- Cardiology Clinic, “St. Spiridon” County Emergency Clinical Hospital, 700111 Iasi, Romania
| | - Elena Rezus
- Department of Rheumatology and Physiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania;
- I Rheumatology Clinic, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa Street, 700661 Iasi, Romania
| | - Ciprian Rezus
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (M.C.B.); (D.P.); (I.I.C.); (C.R.)
- III Internal Medicine Clinic, “St. Spiridon” County Emergency Clinical Hospital, 1 Independence Boulevard, 700111 Iasi, Romania
| |
Collapse
|
11
|
Yang EH, Marmagkiolis K, Balanescu DV, Hakeem A, Donisan T, Finch W, Virmani R, Herrman J, Cilingiroglu M, Grines CL, Toutouzas K, Iliescu C. Radiation-Induced Vascular Disease-A State-of-the-Art Review. Front Cardiovasc Med 2021; 8:652761. [PMID: 33860001 PMCID: PMC8042773 DOI: 10.3389/fcvm.2021.652761] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/05/2021] [Indexed: 12/14/2022] Open
Abstract
Since the 1990s, there has been a steady increase in the number of cancer survivors to an estimated 17 million in 2019 in the US alone. Radiation therapy today is applied to a variety of malignancies and over 50% of cancer patients. The effects of ionizing radiation on cardiac structure and function, so-called radiation-induced heart disease (RIHD), have been extensively studied. We review the available published data on the mechanisms and manifestations of RIHD, with a focus on vascular disease, as well as proposed strategies for its prevention, screening, diagnosis, and management.
Collapse
Affiliation(s)
- Eric H Yang
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | | | - Dinu V Balanescu
- Department of Internal Medicine, Beaumont Hospital, Royal Oak, MI, United States
| | - Abdul Hakeem
- Division of Cardiovascular Diseases & Hypertension, Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Teodora Donisan
- Department of Internal Medicine, Beaumont Hospital, Royal Oak, MI, United States
| | - William Finch
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Renu Virmani
- CVPath Institute, Gaithersburg, MD, United States
| | - Joerg Herrman
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States
| | - Mehmet Cilingiroglu
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,University of Hawaii John Burns School of Medicine, Honolulu, HI, United States
| | - Cindy L Grines
- Cardiovascular Institute, Northside Hospital, Atlanta, GA, United States
| | | | - Cezar Iliescu
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
12
|
Radiobiological Studies of Microvascular Damage through In Vitro Models: A Methodological Perspective. Cancers (Basel) 2021; 13:cancers13051182. [PMID: 33803333 PMCID: PMC7967181 DOI: 10.3390/cancers13051182] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 12/12/2022] Open
Abstract
Ionizing radiation (IR) is used in radiotherapy as a treatment to destroy cancer. Such treatment also affects other tissues, resulting in the so-called normal tissue complications. Endothelial cells (ECs) composing the microvasculature have essential roles in the microenvironment's homeostasis (ME). Thus, detrimental effects induced by irradiation on ECs can influence both the tumor and healthy tissue. In-vitro models can be advantageous to study these phenomena. In this systematic review, we analyzed in-vitro models of ECs subjected to IR. We highlighted the critical issues involved in the production, irradiation, and analysis of such radiobiological in-vitro models to study microvascular endothelial cells damage. For each step, we analyzed common methodologies and critical points required to obtain a reliable model. We identified the generation of a 3D environment for model production and the inclusion of heterogeneous cell populations for a reliable ME recapitulation. Additionally, we highlighted how essential information on the irradiation scheme, crucial to correlate better observed in vitro effects to the clinical scenario, are often neglected in the analyzed studies, limiting the translation of achieved results.
Collapse
|
13
|
Risk of ischaemic cerebrovascular events in head and neck cancer patients is associated with carotid artery radiation dose. Radiother Oncol 2021; 157:182-187. [PMID: 33545259 DOI: 10.1016/j.radonc.2021.01.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 01/13/2021] [Accepted: 01/18/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND AND PURPOSE Radiotherapy in the head and neck area may cause vascular damage to the carotid arteries, increasing the risk of anterior circulation ischaemic cerebrovascular events (ICVEs). However, limited data exists on the relationship between radiation dose to the carotid arteries and risk of ICVE. The purpose of this study was therefore to determine the relationship between radiation dose to the carotid arteries and anterior circulation ICVE risk. MATERIALS AND METHODS A retrospective analysis of a prospective study cohort of 750 head and neck cancer patients treated with definitive (chemo)radiotherapy was performed. Carotid arteries were delineated, and dose-volume parameters of the treatment plans were calculated. ICVEs were scored prospectively and checked retrospectively by analysing all patient records. Cox proportional hazards analysis was performed to analyse the dose-effect relationships. RESULTS The median follow-up period was 3.4 years, 27 patients experienced an ICVE and the 5-year cumulative risk was 4.6%. ICVE risk was significantly associated with dose to the carotid arteries. Multivariable analysis showed that the absolute volume (cm3) of the carotid arteries that received at least a radiation dose of 10 Gy was the most important prognostic factor for ICVE (HR = 1.11, AUC = 0.68, p < 0.001). CONCLUSION This is the first large prospective cohort study that demonstrates an independent dose-effect relationship between radiation dose to the carotid arteries and the risk of ICVE. These findings may be used to identify patients at risk for ICVE after radiotherapy who may benefit from primary or secondary preventive measures.
Collapse
|
14
|
Afifi ANAM, Powerski M, Jechorek D, Brunner TB, Weigt J, Venerito M. Radiation-induced damage in the upper gastrointestinal tract: clinical presentation, diagnostic tests and treatment options. Best Pract Res Clin Gastroenterol 2020; 48-49:101711. [PMID: 33317797 DOI: 10.1016/j.bpg.2020.101711] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/05/2020] [Accepted: 11/09/2020] [Indexed: 01/31/2023]
Abstract
Radiation-induced damage of the upper gastrointestinal (GI) tract results from radiation of GI tumors or structures adjacent to the GI tract. Radiation-induced damages of the upper GI tract may be acute or delayed, and ranges from lack of appetite, mucosal inflammation (i.e. esophagitis, gastritis, duodenitis) to ulcers, which may be complicated by perforation, penetration, bleeding and stenosis. Radiation-related factors as well as individual patient predisposing factors may increase susceptibility to post-radiation damage. High quality evidence for the treatment of radiation-induced GI damage is scarce and the management is often extrapolated from studies on GI lesions of different etiology. Treatment depends on severity and localization of the radiation-induced damage, and ranges from supportive and dietary measures to endoscopic interventions or surgery. Modern radiation techniques may decrease the incidence and severity of the radiation-induced upper gastrointestinal disease.
Collapse
Affiliation(s)
- Ahmed N A M Afifi
- Universitätsklinik für Gastroenterologie, Hepatologie und Infektiologie, Germany
| | - Maciej Powerski
- Universitätsklinik für Radiologie und Nuklearmedizin, Germany
| | | | - Thomas B Brunner
- Universitätsklinik für Strahlentherapie, Otto-von-Guericke Universitätsklinikum Magdeburg, Germany
| | - Jochen Weigt
- Universitätsklinik für Gastroenterologie, Hepatologie und Infektiologie, Germany
| | - Marino Venerito
- Universitätsklinik für Gastroenterologie, Hepatologie und Infektiologie, Germany.
| |
Collapse
|
15
|
Christersdottir T, Pirault J, Gisterå A, Bergman O, Gallina AL, Baumgartner R, Lundberg AM, Eriksson P, Yan ZQ, Paulsson-Berne G, Hansson GK, Olofsson PS, Halle M. Prevention of radiotherapy-induced arterial inflammation by interleukin-1 blockade. Eur Heart J 2020; 40:2495-2503. [PMID: 31081038 PMCID: PMC6685328 DOI: 10.1093/eurheartj/ehz206] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/20/2018] [Accepted: 04/30/2019] [Indexed: 12/15/2022] Open
Abstract
Aims Radiotherapy-induced cardiovascular disease is an emerging problem in a growing population of cancer survivors where traditional treatments, such as anti-platelet and lipid-lowering drugs, have limited benefits. The aim of the study was to investigate vascular inflammatory patterns in human cancer survivors, replicate the findings in an animal model, and evaluate whether interleukin-1 (IL-1) inhibition could be a potential treatment. Methods and results Irradiated human arterial biopsies were collected during microvascular autologous free tissue transfer for cancer reconstruction and compared with non-irradiated arteries from the same patient. A mouse model was used to study the effects of the IL-1 receptor antagonist, anakinra, on localized radiation-induced vascular inflammation. We observed significant induction of genes associated with inflammasome biology in whole transcriptome analysis of irradiated arteries, a finding supported by elevated protein levels in irradiated arteries of both, pro-caspase and caspase-1. mRNA levels of inflammasome associated chemokines CCL2, CCL5 together with the adhesion molecule VCAM1, were elevated in human irradiated arteries as was the number of infiltrating macrophages. A similar pattern was reproduced in Apoe−/− mouse 10 weeks after localized chest irradiation with 14 Gy. Treatment with anakinra in irradiated mice significantly reduced Ccl2 and Ccl5 mRNA levels and expression of I-Ab. Conclusion Anakinra, administered directly after radiation exposure for 2 weeks, ameliorated radiation induced sustained expression of inflammatory mediators in mice. Further studies are needed to evaluate IL-1 blockade as a treatment of radiotherapy-induced vascular disease in a clinical setting. ![]()
Collapse
Affiliation(s)
- Tinna Christersdottir
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.,St. Erik Eye Hospital, Stockholm, Sweden
| | - John Pirault
- Cardiovascular Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Bioclinicum J8:20, Visionsgatan 4, Stockholm, Sweden
| | - Anton Gisterå
- Cardiovascular Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Bioclinicum J8:20, Visionsgatan 4, Stockholm, Sweden
| | - Otto Bergman
- Cardiovascular Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Bioclinicum J8:20, Visionsgatan 4, Stockholm, Sweden
| | - Alessandro L Gallina
- Cardiovascular Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Bioclinicum J8:20, Visionsgatan 4, Stockholm, Sweden
| | - Roland Baumgartner
- Cardiovascular Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Bioclinicum J8:20, Visionsgatan 4, Stockholm, Sweden
| | - Anna M Lundberg
- Cardiovascular Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Bioclinicum J8:20, Visionsgatan 4, Stockholm, Sweden
| | - Per Eriksson
- Cardiovascular Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Bioclinicum J8:20, Visionsgatan 4, Stockholm, Sweden
| | - Zhong-Qun Yan
- Cardiovascular Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Bioclinicum J8:20, Visionsgatan 4, Stockholm, Sweden
| | - Gabrielle Paulsson-Berne
- Cardiovascular Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Bioclinicum J8:20, Visionsgatan 4, Stockholm, Sweden
| | - Göran K Hansson
- Cardiovascular Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Bioclinicum J8:20, Visionsgatan 4, Stockholm, Sweden
| | - Peder S Olofsson
- Cardiovascular Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital, Bioclinicum J8:20, Visionsgatan 4, Stockholm, Sweden
| | - Martin Halle
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.,Reconstructive Plastic Surgery, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
16
|
Obrador E, Salvador R, Villaescusa JI, Soriano JM, Estrela JM, Montoro A. Radioprotection and Radiomitigation: From the Bench to Clinical Practice. Biomedicines 2020; 8:E461. [PMID: 33142986 PMCID: PMC7692399 DOI: 10.3390/biomedicines8110461] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 02/07/2023] Open
Abstract
The development of protective agents against harmful radiations has been a subject of investigation for decades. However, effective (ideal) radioprotectors and radiomitigators remain an unsolved problem. Because ionizing radiation-induced cellular damage is primarily attributed to free radicals, radical scavengers are promising as potential radioprotectors. Early development of such agents focused on thiol synthetic compounds, e.g., amifostine (2-(3-aminopropylamino) ethylsulfanylphosphonic acid), approved as a radioprotector by the Food and Drug Administration (FDA, USA) but for limited clinical indications and not for nonclinical uses. To date, no new chemical entity has been approved by the FDA as a radiation countermeasure for acute radiation syndrome (ARS). All FDA-approved radiation countermeasures (filgrastim, a recombinant DNA form of the naturally occurring granulocyte colony-stimulating factor, G-CSF; pegfilgrastim, a PEGylated form of the recombinant human G-CSF; sargramostim, a recombinant granulocyte macrophage colony-stimulating factor, GM-CSF) are classified as radiomitigators. No radioprotector that can be administered prior to exposure has been approved for ARS. This differentiates radioprotectors (reduce direct damage caused by radiation) and radiomitigators (minimize toxicity even after radiation has been delivered). Molecules under development with the aim of reaching clinical practice and other nonclinical applications are discussed. Assays to evaluate the biological effects of ionizing radiations are also analyzed.
Collapse
Affiliation(s)
- Elena Obrador
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (E.O.); (R.S.); (J.M.E.)
| | - Rosario Salvador
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (E.O.); (R.S.); (J.M.E.)
| | - Juan I. Villaescusa
- Service of Radiological Protection, Clinical Area of Medical Image, La Fe University Hospital, 46026 Valencia, Spain;
- Biomedical Imaging Research Group GIBI230, Health Research Institute (IISLaFe), La Fe University Hospital, 46026 Valencia, Spain
| | - José M. Soriano
- Food & Health Lab, Institute of Materials Science, University of Valencia, 46980 Valencia, Spain;
- Joint Research Unit in Endocrinology, Nutrition and Clinical Dietetics, University of Valencia-Health Research Institute IISLaFe, 46026 Valencia, Spain
| | - José M. Estrela
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (E.O.); (R.S.); (J.M.E.)
| | - Alegría Montoro
- Service of Radiological Protection, Clinical Area of Medical Image, La Fe University Hospital, 46026 Valencia, Spain;
- Biomedical Imaging Research Group GIBI230, Health Research Institute (IISLaFe), La Fe University Hospital, 46026 Valencia, Spain
| |
Collapse
|
17
|
Jang H, Kwak SY, Park S, Kim K, Kim YH, Na J, Kim H, Jang WS, Lee SJ, Kim MJ, Myung JK, Shim S. Pravastatin Alleviates Radiation Proctitis by Regulating Thrombomodulin in Irradiated Endothelial Cells. Int J Mol Sci 2020; 21:ijms21051897. [PMID: 32164317 PMCID: PMC7084904 DOI: 10.3390/ijms21051897] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/07/2020] [Accepted: 03/08/2020] [Indexed: 12/23/2022] Open
Abstract
Although radiotherapy plays a crucial in the management of pelvic tumors, its toxicity on surrounding healthy tissues such as the small intestine, colon, and rectum is one of the major limitations associated with its use. In particular, proctitis is a major clinical complication of pelvic radiotherapy. Recent evidence suggests that endothelial injury significantly affects the initiation of radiation-induced inflammation. The damaged endothelial cells accelerate immune cell recruitment by activating the expression of endothelial adhesive molecules, which participate in the development of tissue damage. Pravastatin, a cholesterol lowering drug, exerts persistent anti-inflammatory and anti-thrombotic effects on irradiated endothelial cells and inhibits the interaction of leukocytes and damaged endothelial cells. Here, we aimed to investigate the effects of pravastatin on radiation-induced endothelial damage in human umbilical vein endothelial cell and a murine proctitis model. Pravastatin attenuated epithelial damage and inflammatory response in irradiated colorectal lesions. In particular, pravastatin improved radiation-induced endothelial damage by regulating thrombomodulin (TM) expression. In addition, exogenous TM inhibited leukocyte adhesion to the irradiated endothelial cells. Thus, pravastatin can inhibit endothelial damage by inducing TM, thereby alleviating radiation proctitis. Therefore, we suggest that pharmacological modulation of endothelial TM may limit intestinal inflammation after irradiation.
Collapse
Affiliation(s)
- Hyosun Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
| | - Seo-Young Kwak
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
| | - Sunhoo Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
- Department of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea
| | - Kyuchang Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
| | - Young-heon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
| | - Jiyoung Na
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
| | - Hyewon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
| | - Won-Suk Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
| | - Sun-Joo Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
| | - Min Jung Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
| | - Jae Kyung Myung
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
- Department of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea
| | - Sehwan Shim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (H.J.); (S.-Y.K.); (S.P.); (K.K.); (Y.-h.K.); (J.N.); (H.K.); (W.-S.J.); (S.-J.L.); (M.J.K.); (J.K.M.)
- Correspondence: ; Tel.: +82-2-3399-5873
| |
Collapse
|
18
|
Bykov VN, Grebenyuk AN, Ushakov IB. The Use of Radioprotective Agents to Prevent Effects Associated with Aging. BIOL BULL+ 2019. [DOI: 10.1134/s1062359019120021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
19
|
Jones JW, Alloush J, Sellamuthu R, Chua HL, MacVittie TJ, Orschell CM, Kane MA. Effect of Sex on Biomarker Response in a Mouse Model of the Hematopoietic Acute Radiation Syndrome. HEALTH PHYSICS 2019; 116:484-502. [PMID: 30681425 PMCID: PMC6384137 DOI: 10.1097/hp.0000000000000961] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Sex is an important confounding variable in biomarker development that must be incorporated into biomarker discovery and validation. Additionally, understanding of sex as a biological variable is essential for effective translation of biomarkers in animal models to human populations. Toward these ends, we conducted high-throughput targeted metabolomics using liquid chromatography tandem mass spectrometry and multiplexed immunoassay analyses using a Luminex-based system in both male and female mice in a model of total-body irradiation at a radiation dose consistent with the hematopoietic acute radiation syndrome. Metabolomic and immunoassay analyses identified metabolites and cytokines that were significantly different in plasma from naive and irradiated C57BL/6 mice consisting of equal numbers of female and male mice at 3 d after 8.0 or 8.72 Gy, an approximate LD60-70/30 dose of total-body irradiation. An additional number of metabolites and cytokines had sex-specific responses after radiation. Analyses of sham-irradiated mice illustrate the presence of stress-related changes in several cytokines due simply to undergoing the irradiation procedure, absent actual radiation exposure. Basal differences in metabolite levels between female and male were also identified as well as time-dependent changes in cytokines up to 9 d postexposure. These studies provide data toward defining the influence of sex on plasma-based biomarker candidates in a well-defined mouse model of acute radiation syndrome.
Collapse
Affiliation(s)
- Jace W. Jones
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| | - Jenna Alloush
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| | | | - Hui Lin Chua
- Indiana University School of Medicine, Indianapolis, IN
| | - Thomas J. MacVittie
- University of Maryland, School of Medicine, Department of Radiation Oncology, Baltimore, MD
| | | | - Maureen A. Kane
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD
| |
Collapse
|
20
|
Balog RP, Bacher R, Chang P, Greenstein M, Jammalamadaka S, Javitz H, Knox SJ, Lee S, Lin H, Shaler T, Shura L, Stein P, Todd K, Cooper DE. Development of a biodosimeter for radiation triage using novel blood protein biomarker panels in humans and non-human primates. Int J Radiat Biol 2019; 96:22-34. [PMID: 30605362 DOI: 10.1080/09553002.2018.1532611] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Purpose: In a significant nuclear event, hundreds of thousands of individuals will require rapid triage for absorbed radiation to ensure effective medical treatment and efficient use of medical resources. We are developing a rapid screening method to assess whether an individual received an absorbed dose of ≥2 Gy based on the analysis of a specific panel of blood proteins in a fingerstick blood sample.Materials and methods: We studied a data set of 1051 human blood samples obtained from radiotherapy patients, normal healthy individuals, and several special population groups. We compared the findings in humans with those from irradiation studies in non-human primates (NHPs).Results: We identified a panel of three protein biomarkers, salivary alpha amylase (AMY1), Flt3 ligand (FLT3L), and monocyte chemotactic protein 1 (MCP1), which are upregulated in human patients receiving fractionated doses of total body irradiation (TBI) therapy as a treatment for cancer. These proteins exhibited a similar radiation response in NHPs after single acute or fractionated doses of ionizing radiation.Conclusion: Our work provides confidence in this biomarker panel for biodosimetry triage using fingerstick blood samples and in the use of NHPs as a model for irradiated humans.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Susan J Knox
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | | | - Hua Lin
- SRI International, Menlo Park, CA, USA
| | | | - Lei Shura
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | | | | | | |
Collapse
|
21
|
Jang H, Lee J, Park S, Myung H, Kang J, Kim K, Kim H, Jang WS, Lee SJ, Shim S, Myung JK. Pravastatin Attenuates Acute Radiation-Induced Enteropathy and Improves Epithelial Cell Function. Front Pharmacol 2018; 9:1215. [PMID: 30459609 PMCID: PMC6232864 DOI: 10.3389/fphar.2018.01215] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/05/2018] [Indexed: 12/12/2022] Open
Abstract
Background and Aim: Radiation-induced enteropathy is frequently observed after radiation therapy for abdominal and pelvic cancer or occurs secondary to accidental radiation exposure. The acute effects of irradiation on the intestine might be attributed to inhibition of mitosis in the crypts, as the loss of proliferative functions impairs development of the small intestinal epithelium and its barrier function. Especially, oxidative damage to intestinal epithelial cells is a key event in the initiation and progression of radiation-induced enteropathy. Pravastatin is widely used clinically to lower serum cholesterol levels and has been reported to have anti-inflammatory effects on endothelial cells. Here, we investigated the therapeutic effects of pravastatin on damaged epithelial cells after radiation-induced enteritis using in vitro and in vivo systems. Materials and Methods: To evaluate the effects of pravastatin on intestinal epithelial cells, we analyzed proliferation and senescence, oxidative damage, and inflammatory cytokine expression in an irradiated human intestinal epithelial cell line (InEpC). In addition, to investigate the therapeutic effects of pravastatin in mice, we performed histological analysis, bacterial translocation assays, and intestinal permeability assays, and also assessed inflammatory cytokine expression, using a radiation-induced enteropathy model. Results: Histological damage such as shortening of villi length and impaired intestinal crypt function was observed in whole abdominal-irradiated mice. However, damage was attenuated in pravastatin-treated animals, in which normalization of intestinal epithelial cell differentiation was also observed. Using in vitro and in vivo systems, we also showed that pravastatin improves the proliferative properties of intestinal epithelial cells and decreases radiation-induced oxidative damage to the intestine. In addition, pravastatin inhibited levels of epithelial-derived inflammatory cytokines including IL-6, IL-1β, and TNF-α in irradiated InEpC cells. We also determined that pravastatin could rescue intestinal barrier dysfunction via anti-inflammatory effects using the mouse model. Conclusion: Pravastatin has a therapeutic effect on intestinal lesions and attenuates radiation-induced epithelial damage by suppressing oxidative stress and the inflammatory response.
Collapse
Affiliation(s)
- Hyosun Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Janet Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Sunhoo Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea.,Department of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Hyunwook Myung
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Jihoon Kang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Kyuchang Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Hyewon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Won-Suk Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Sun-Joo Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Sehwan Shim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| | - Jae K Myung
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea.,Department of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, South Korea
| |
Collapse
|
22
|
Upregulation of Plasminogen Activator Inhibitor-1 in Irradiated Recipient Arteries and Veins from Free Tissue Transfer Reconstruction in Cancer Patients. Mediators Inflamm 2018; 2018:4058986. [PMID: 30402041 PMCID: PMC6193344 DOI: 10.1155/2018/4058986] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 07/30/2018] [Indexed: 12/14/2022] Open
Abstract
Background Clinical studies have shown that radiotherapy can induce vascular disease at the site of exposure but is usually not clinically evident until years after treatment. We have studied irradiated human arteries and veins to better understand the underlying biology in search of future treatments. The aim was to investigate whether radiotherapy contributed to a sustained expression of plasminogen activator inhibitor-1 (PAI-1) in human arteries and veins. Methods Irradiated arteries and veins were harvested, together with unirradiated control vessels, from patients undergoing free tissue transfer reconstruction at a median time of 90 weeks [5–650] following radiation exposure. Differential gene expression of PAI-1 was analysed, together with immunohistochemistry (IHC) and immunofluorescence (IF). Results PAI-1 gene expression was increased in both arteries (p = 0.012) and veins (p < 0.001) in irradiated compared to unirradiated control vessels. IHC and IF indicated that cells expressing PAI-1 were located in the adventitia of both arteries and veins and colocalized with cells positive for CD68, CD45, and α-SMA in arteries and with CD45 and α-SMA in veins. Conclusion The current study shows a sustained upregulation of PAI-1 in both arteries and veins after exposure to ionizing radiation, indicating a chronic inflammation mainly in the adventitia. We believe that the results contribute to further understanding of radiation-induced vascular disease, where targeting PAI-1 may be a potential treatment.
Collapse
|
23
|
Radiation-induced muscle fibrosis rat model: establishment and valuation. Radiat Oncol 2018; 13:160. [PMID: 30157899 PMCID: PMC6114061 DOI: 10.1186/s13014-018-1104-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 08/17/2018] [Indexed: 02/06/2023] Open
Abstract
Background Lack of animal model of radiation induced muscle fibrosis, this study aimed to establish such a model by using 90 Gy single dose irradiation to mimic clinical relevance and also to explore the potential post-irradiation regenerative mechanism. Methods SD rats were randomly divided into dose investigation groups and time gradient groups. Group1–6 were irradiated with a single dose of 65Gy, 70Gy, 75Gy, 80Gy, 85Gy and 90Gy respectively, and the degree of rectus femoris fibrosis in the irradiated area was detected at 4 weeks after irradiation. Group 7–9 were irradiated with a single dose of 90Gy, and the results were detected 1, 2, 4, and 8 weeks after irradiation. Then the general condition of rats was recorded. Masson staining was used to detect muscle fibrosis. The ultrastructure of muscles was observed by electron microscope, and the expression changes of satellite cell proliferation and differentiation related genes were detected by quantitative real-time-PCR. Results A single dose of 90Gy irradiation could cause muscle fibrosis in rats. As time goes on, the severity of muscle fibrosis and the expression of TGF- β1 increased. Significant swelling of mitochondria, myofilament disarrangement and dissolution, obvious endothelial cell swelling, increased vascular permeability, decrease of blood cell, deposition of fibrosis tissue around the vessel could be found compared with the control group. At around the 4th week, the expressions of Pax7, Myf5, MyoD, MyoG, Mrf4 increased. Conclusion Irradiation of 90Gy can successfully establish the rat model of radiation-induced muscle fibrosis. This model demonstrated that regenerative process was initiated by the irradiation only at an early stage, which can serve a suitable model for investigating regenerative therapy for post-radiation muscle fibrosis.
Collapse
|
24
|
Hui L, Shijun H, Tao L, Guoqiang W, Shixiong H. Bilateral thalamic and mesencephalic infarctions with hypopituitarism as long-term complications postradiotherapy: A case report. Medicine (Baltimore) 2018; 97:e11917. [PMID: 30142801 PMCID: PMC6113035 DOI: 10.1097/md.0000000000011917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Radiation is widely used as the first-line treatment for nasopharyngeal carcinoma (NPC) and improves survival. Nevertheless, radiation also places the patients at risk of radiation-induced adverse effects, such as transient ischemic attack, ischemic stroke, hypopituitarism, and cranial nerve and temporal lobe dysfunction. CASE REPORT A 54-year-old woman who had undergone radiation treatment for NPC 14 years earlier and had no cerebrovascular risk factors, visited our department 4 days after sudden onset of consciousness disturbance. Brain magnetic resonance imaging (MRI) revealed bilateral thalamic and left mesencephalic infarctions with empty sella. Meanwhile, MR angiography showed narrowing in the bilateral posterior cerebral artery. Furthermore, laboratory tests showed low total triiodothyronine (T3), thyroxine (T4), free T3, free T4, luteinizing hormone, estradiol, follicle-stimulating hormone, and serum natrium and normal thyroid-stimulating hormone, which indicated radiation-related hypopituitarism. Serologically, she had low hemoglobin, hematocrit, mean corpuscular volume, mean corpuscular hemoglobin, mean corpuscular hemoglobin concentration, ferritin, and serum iron levels and elevated transferrin, manifesting microcytic anemia. The treatment, including aspirin, atorvastatin, levothyroxine, prednisone, saline infusion, and chalybeate, promoted the patient's recovery. CONCLUSION To our knowledge, this is the first report of bilateral thalamic and mesencephalic infarction together with hypopituitarism following radiotherapy for NPC.
Collapse
|
25
|
Nguyen HQ, To NH, Zadigue P, Kerbrat S, De La Taille A, Le Gouvello S, Belkacemi Y. Ionizing radiation-induced cellular senescence promotes tissue fibrosis after radiotherapy. A review. Crit Rev Oncol Hematol 2018; 129:13-26. [PMID: 30097231 DOI: 10.1016/j.critrevonc.2018.06.012] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 06/08/2018] [Accepted: 06/15/2018] [Indexed: 02/06/2023] Open
Abstract
Ionizing radiation-exposure induces a variety of cellular reactions, such as senescence and apoptosis. Senescence is a permanent arrest state of the cell division, which can be beneficial or detrimental for normal tissue via an inflammatory response and senescence-associated secretion phenotype. Damage to healthy cells and their microenvironment is considered as an important source of early and late complications with an increased risk of morbidity in patients after radiotherapy (RT). In addition, the benefit/risk ratio may depend on the radiation technique/dose used for cancer eradication and the irradiated volume of healthy tissues. For radiation-induced fibrosis risk, the knowledge of mechanisms and potential prevention has become a crucial point to determining radiation parameters and patients' intrinsic radiosensitivity. This review summarizes our understanding of ionizing radiation-induced senescent cell in fibrogenesis. This mechanism may provide new insights for therapeutic modalities for better risk/benefit ratios after RT in the new era of personalized treatments.
Collapse
Affiliation(s)
- Hoang Quy Nguyen
- University of Paris Saclay, University of Paris Est Créteil (UPEC), France, University of Medicine and Pharmacy, Ho Chi Minh City, Viet Nam; INSERM U955 Team 07, Créteil, France
| | - Nhu Hanh To
- INSERM U955 Team 07, Créteil, France; APHP, Department of Radiation Oncology and Henri Mondor Breast Cancer and, Henri Mondor University Hospital, University of Paris Est Créteil (UPEC), France
| | | | - Stéphane Kerbrat
- INSERM U955 Team 04, University of Paris Est Créteil (UPEC), France
| | - Alexandre De La Taille
- INSERM U955 Team 07, Créteil, France; APHP, Department of Urology, Henri Mondor University Hospital, University of Paris Est Créteil (UPEC), Créteil, France
| | - Sabine Le Gouvello
- INSERM U955 Team 04, University of Paris Est Créteil (UPEC), France; APHP, Department of Biology & Pathology, Henri Mondor University Hospital, University of Paris Est Créteil (UPEC), Créteil, France
| | - Yazid Belkacemi
- INSERM U955 Team 07, Créteil, France; APHP, Department of Radiation Oncology and Henri Mondor Breast Cancer and, Henri Mondor University Hospital, University of Paris Est Créteil (UPEC), France.
| |
Collapse
|
26
|
Fernández-Alvarez V, López F, Suárez C, Strojan P, Eisbruch A, Silver CE, Mendenhall WM, Langendijk JA, Rinaldo A, Lee AWM, Beitler JJ, Smee R, Alvarez J, Ferlito A. Radiation-induced carotid artery lesions. Strahlenther Onkol 2018; 194:699-710. [DOI: 10.1007/s00066-018-1304-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 04/10/2018] [Indexed: 11/24/2022]
|
27
|
Najafi M, Motevaseli E, Shirazi A, Geraily G, Rezaeyan A, Norouzi F, Rezapoor S, Abdollahi H. Mechanisms of inflammatory responses to radiation and normal tissues toxicity: clinical implications. Int J Radiat Biol 2018; 94:335-356. [PMID: 29504497 DOI: 10.1080/09553002.2018.1440092] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 01/23/2018] [Accepted: 01/31/2018] [Indexed: 02/06/2023]
Abstract
PURPOSE Cancer treatment is one of the most challenging diseases in the present era. Among a few modalities for cancer therapy, radiotherapy plays a pivotal role in more than half of all treatments alone or combined with other cancer treatment modalities. Management of normal tissue toxicity induced by radiation is one of the most important limiting factors for an appropriate radiation treatment course. The evaluation of mechanisms of normal tissue toxicity has shown that immune responses especially inflammatory responses play a key role in both early and late side effects of exposure to ionizing radiation (IR). DNA damage and cell death, as well as damage to some organelles such as mitochondria initiate several signaling pathways that result in the response of immune cells. Massive cell damage which is a common phenomenon following exposure to a high dose of IR cause secretion of a lot of inflammatory mediators including cytokines and chemokines. These mediators initiate different changes in normal tissues that may continue for a long time after irradiation. In this study, we reviewed the mechanisms of inflammatory responses to IR that are involved in normal tissue toxicity and considered as the most important limiting factors in radiotherapy. Also, we introduced some agents that have been proposed for management of these responses. CONCLUSIONS The early inflammation during the radiation treatment is often a limiting factor in radiotherapy. In addition to the limiting factors, chronic inflammatory responses may increase the risk of second primary cancers through continuous free radical production, attenuation of tumor suppressor genes, and activation of oncogenes. Moreover, these effects may influence non-irradiated tissues through a mechanism named bystander effect.
Collapse
Affiliation(s)
- Masoud Najafi
- a Radiology and Nuclear Medicine Department, School of Paramedical Sciences , Kermanshah University of Medical Science , Kermanshah , Iran
| | - Elahe Motevaseli
- b Department of Molecular Medicine, School of Advanced Technologies in Medicine , Tehran University of Medical Sciences , Tehran , Iran
| | - Alireza Shirazi
- c Department of Medical Physics and Biomedical Engineering, Faculty of Medicine , Tehran University of Medical Sciences , Tehran , Iran
| | - Ghazale Geraily
- c Department of Medical Physics and Biomedical Engineering, Faculty of Medicine , Tehran University of Medical Sciences , Tehran , Iran
| | - Abolhasan Rezaeyan
- d Department of Medical Physics, School of Medicine , Iran University of Medical Sciences , Tehran , Iran
| | - Farzad Norouzi
- e Science and Research Branch , Azad University , Tehran , Iran
| | - Saeed Rezapoor
- f Department of Radiology, Faculty of Paramedical Sciences , Tehran University of Medical Sciences , Tehran , Iran
| | - Hamid Abdollahi
- d Department of Medical Physics, School of Medicine , Iran University of Medical Sciences , Tehran , Iran
| |
Collapse
|
28
|
Addison D, Lawler PR, Emami H, Janjua SA, Staziaki PV, Hallett TR, Hennessy O, Lee H, Szilveszter B, Lu M, Mousavi N, Nayor MG, Delling FN, Romero JM, Wirth LJ, Chan AW, Hoffmann U, Neilan TG. Incidental Statin Use and the Risk of Stroke or Transient Ischemic Attack after Radiotherapy for Head and Neck Cancer. J Stroke 2018; 20:71-79. [PMID: 29402065 PMCID: PMC5836583 DOI: 10.5853/jos.2017.01802] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/01/2017] [Accepted: 12/05/2017] [Indexed: 12/29/2022] Open
Abstract
Background and Purpose Interventions to reduce the risk for cerebrovascular events (CVE; stroke and transient ischemic attack [TIA]) after radiotherapy (RT) for head and neck cancer (HNCA) are needed. Among broad populations, statins reduce CVEs; however, whether statins reduce CVEs after RT for HNCA is unclear. Therefore, we aimed to test whether incidental statin use at the time of RT is associated with a lower rate of CVEs after RT for HNCA. Methods From an institutional database we identified all consecutive subjects treated with neck RT from 2002 to 2012 for HNCA. Data collection and event adjudication was performed by blinded teams. The primary outcome was a composite of ischemic stroke and TIA. The secondary outcome was ischemic stroke. The association between statin use and events was determined using Cox proportional hazard models after adjustment for traditional and RT-specific risk factors. Results The final cohort consisted of 1,011 patients (59±13 years, 30% female, 44% hypertension) with 288 (28%) on statins. Over a median follow-up of 3.4 years (interquartile range, 0.1 to 14) there were 102 CVEs (89 ischemic strokes and 13 TIAs) with 17 in statin users versus 85 in nonstatins users. In a multivariable model containing known predictors of CVE, statins were associated with a reduction in the combination of stroke and TIA (hazard ratio [HR], 0.4; 95% confidence interval [CI], 0.2 to 0.8; P=0.01) and ischemic stroke alone (HR, 0.4; 95% CI, 0.2 to 0.8; P=0.01). Conclusions Incidental statin use at the time of RT for HNCA is associated with a lower risk of stroke or TIA.
Collapse
Affiliation(s)
- Daniel Addison
- Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Division of Cardiology, Department of Medicine, The Ohio State University, Columbus, OH, USA
| | - Patrick R Lawler
- Peter Munk Cardiac Centre, Toronto General Hospital, and the Heart and Stroke Richard Lewar Centre of Excellence, University of Toronto, Toronto, ON, Canada.,Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hamed Emami
- Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sumbal A Janjua
- Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Pedro V Staziaki
- Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Travis R Hallett
- Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Orla Hennessy
- Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hang Lee
- Biostatistics Center, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Bálint Szilveszter
- Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael Lu
- Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Negar Mousavi
- Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthew G Nayor
- Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Francesca N Delling
- Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Javier M Romero
- Division of Neuroradiology, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lori J Wirth
- Division of Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Annie W Chan
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Udo Hoffmann
- Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tomas G Neilan
- Cardiac MR PET CT Program, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
29
|
Kim BJ, Kang HG, Lee SW, Jung J, Lee MH, Kang DW, Kim JS, Kwon SU. Changes in the Common Carotid Artery after Radiotherapy: Wall Thickness, Calcification, and Atherosclerosis. J Clin Neurol 2018; 14:35-42. [PMID: 29629538 PMCID: PMC5765254 DOI: 10.3988/jcn.2018.14.1.35] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 08/25/2017] [Accepted: 08/29/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Since the long-term survival rate has improved in laryngeal cancer patients who receive radiotherapy, concerns about postradiation complications (including carotid atherosclerosis) have increased. We followed changes in the common carotid artery (CCA) after radiotherapy and identified the underlying risk factors. METHODS Consecutive patients with laryngeal cancer who underwent radiotherapy between January 1999 and December 2009 and who had received computed tomography (CT) both pre- and postradiotherapy were enrolled. Changes in the wall thickness and in the vessel and lumen areas as well as the presence of calcification or atherosclerosis were investigated. Demographics and risk factors were compared between patients with and without atherosclerosis at follow-up CT. RESULTS In total, 125 patients were enrolled. The wall thickness had increased and the lumen area had decreased several months after radiotherapy. These changes were not associated with vascular risk factors and were not progressive. Calcification and atherosclerosis were observed in 37 (29.6%) and 71 (56.8%) patients, respectively. Diabetes was associated with calcification (p=0.02). The prevalence of hyperlipidemia was higher in patients with atherosclerosis (28.2% vs. 11.1%, p=0.02) and for a longer period postradiation [62.7±32.1 vs. 40.0±24.2 months (mean±SD), p<0.001]. Atherosclerosis occurred mostly in the middle portion of the CCA (n=31, 24.6%), followed by the proximal CCA at the intrathoracic level (n=26, 20.6%) and the distal CCA (n=6, 4.8%). Positive remodeling was also observed, but this was less common in patients with calcification (p=0.02). CONCLUSIONS Various types of postradiation changes occur in the CCA and can be easily observed in postradiation CT. The prevalence and burden of postradiation atherosclerosis increased in a close relationship with baseline cholesterol levels and the time after radiotherapy. Postradiation atherosclerosis was observed at unusual sites of the CCA.
Collapse
Affiliation(s)
- Bum Joon Kim
- Department of Neurology, Kyung Hee University Hospital, Kyung Hee University School of Medicine, Seoul, Korea
| | - Hyun Goo Kang
- Department of Neurology, Chosun University Hospital, Gwangju, Korea
| | - Sang Wook Lee
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jinhong Jung
- Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Min Hwan Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Dong Wha Kang
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jong S Kim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sun U Kwon
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
30
|
Naeimi RA, Talebpour Amiri F, Khalatbary AR, Ghasemi A, Zargari M, Ghesemi M, Hosseinimehr SJ. Atorvastatin mitigates testicular injuries induced by ionizing radiation in mice. Reprod Toxicol 2017; 72:115-121. [DOI: 10.1016/j.reprotox.2017.06.052] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 06/14/2017] [Accepted: 06/18/2017] [Indexed: 01/13/2023]
|
31
|
Rho inhibition by lovastatin affects apoptosis and DSB repair of primary human lung cells in vitro and lung tissue in vivo following fractionated irradiation. Cell Death Dis 2017; 8:e2978. [PMID: 28796249 PMCID: PMC5596560 DOI: 10.1038/cddis.2017.372] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 06/22/2017] [Accepted: 07/02/2017] [Indexed: 12/12/2022]
Abstract
Thoracic radiotherapy causes damage of normal lung tissue, which limits the cumulative radiation dose and, hence, confines the anticancer efficacy of radiotherapy and impacts the quality of life of tumor patients. Ras-homologous (Rho) small GTPases regulate multiple stress responses and cell death. Therefore, we investigated whether pharmacological targeting of Rho signaling by the HMG-CoA-reductase inhibitor lovastatin influences ionizing radiation (IR)-induced toxicity in primary human lung fibroblasts, lung epithelial and lung microvascular endothelial cells in vitro and subchronic mouse lung tissue damage following hypo-fractionated irradiation (4x4 Gy). The statin improved the repair of radiation-induced DNA double-strand breaks (DSBs) in all cell types and, moreover, protected lung endothelial cells from IR-induced caspase-dependent apoptosis, likely involving p53-regulated mechanisms. Under the in vivo situation, treatment with lovastatin or the Rac1-specific small molecule inhibitor EHT1864 attenuated the IR-induced increase in breathing frequency and reduced the percentage of γH2AX and 53BP1-positive cells. This indicates that inhibition of Rac1 signaling lowers IR-induced residual DNA damage by promoting DNA repair. Moreover, lovastatin and EHT1864 protected lung tissue from IR-triggered apoptosis and mitigated the IR-stimulated increase in regenerative proliferation. Our data document beneficial anti-apoptotic and genoprotective effects of pharmacological targeting of Rho signaling following hypo-fractionated irradiation of lung cells in vitro and in vivo. Rac1-targeting drugs might be particular useful for supportive care in radiation oncology and, moreover, applicable to improve the anticancer efficacy of radiotherapy by widening the therapeutic window of thoracic radiation exposure.
Collapse
|
32
|
Serrano NA, Kalman NS, Anscher MS. Reducing rectal injury in men receiving prostate cancer radiation therapy: current perspectives. Cancer Manag Res 2017; 9:339-350. [PMID: 28814898 PMCID: PMC5546182 DOI: 10.2147/cmar.s118781] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Dose escalation is now the standard of care for the treatment of prostate cancer with radiation therapy. However, the rectum tends to be the dose-limiting structure when treating prostate cancer, given its close proximity. Early and late toxicities can occur when the rectum receives large doses of radiation therapy. New technologies allow for prevention of these toxicities. In this review, we examine the evidence that supports various dose constraints employed to prevent these rectal injuries from occurring. We also examine the use of intensity-modulated radiation therapy and how this compares to older radiation therapy techniques that allow for further sparing of the rectum during a radiation therapy course. We then review the literature on endorectal balloons and the effects of their daily use throughout a radiation therapy course. Tissue spacers are now being investigated in greater detail; these devices are injected into the rectoprostatic fascia to physically increase the distance between the prostate and the anterior rectal wall. Last, we review the use of systemic drugs, specifically statin medications and antihypertensives, as well as their impact on rectal toxicity.
Collapse
Affiliation(s)
- Nicholas A Serrano
- Department of Radiation Oncology, Virginia Commonwealth University - Massey Cancer Center, Richmond, VA
| | - Noah S Kalman
- Department of Radiation Oncology, Virginia Commonwealth University - Massey Cancer Center, Richmond, VA
| | - Mitchell S Anscher
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
33
|
McLaughlin MF, Donoviel DB, Jones JA. Novel Indications for Commonly Used Medications as Radiation Protectants in Spaceflight. Aerosp Med Hum Perform 2017. [PMID: 28641684 DOI: 10.3357/amhp.4735.2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND In the space environment, the traditional radioprotective principles of time, distance, and shielding become difficult to implement. Additionally, the complex radiation environment inherent in space, the chronic exposure timeframe, and the presence of numerous confounding variables complicate the process of creating appropriate risk models for astronaut exposure. Pharmaceutical options hold tremendous promise to attenuate acute and late effects of radiation exposure in the astronaut population. Pharmaceuticals currently approved for other indications may also offer radiation protection, modulation, or mitigation properties along with a well-established safety profile. Currently there are only three agents which have been clinically approved to be employed for radiation exposure, and these only for very narrow indications. This review identifies a number of agents currently approved by the U.S. Food and Drug Administration (FDA) which could warrant further investigation for use in astronauts. Specifically, we examine preclinical and clinical evidence for statins, nonsteroidal anti-inflammatory drugs (NSAIDs), angiotensin converting enzyme inhibitors (ACEIs), angiotensin II receptor blockers (ARBs), metformin, calcium channel blockers, β adrenergic receptor blockers, fingolimod, N-acetylcysteine, and pentoxifylline as potential radiation countermeasures.McLaughlin MF, Donoviel DB, Jones JA. Novel indications for commonly used medications as radiation protectants in spaceflight. Aerosp Med Hum Perform. 2017; 88(7):665-676.
Collapse
|
34
|
Lemanska A, Byford RC, Correa A, Cruickshank C, Dearnaley DP, Griffin C, Hall E, de Lusignan S, Faithfull S. Linking CHHiP prostate cancer RCT with GP records: A study proposal to investigate the effect of co-morbidities and medications on long-term symptoms and radiotherapy-related toxicity. Tech Innov Patient Support Radiat Oncol 2017; 2:5-12. [PMID: 32095558 PMCID: PMC7033766 DOI: 10.1016/j.tipsro.2017.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/17/2017] [Accepted: 06/07/2017] [Indexed: 12/25/2022] Open
Abstract
Background Patients receiving cancer treatment often have one or more co-morbid conditions that are treated pharmacologically. Co-morbidities are recorded in clinical trials usually only at baseline. However, co-morbidities evolve and new ones emerge during cancer treatment. The interaction between multi-morbidity and cancer recovery is significant but poorly understood. Purpose To investigate the effect of co-morbidities (e.g. cardiovascular and diabetes) and medications (e.g. statins, antihypertensives, metformin) on radiotherapy-related toxicity and long-term symptoms in order to identify potential risk factors. The possible protective effect of medications such as statins or antihypertensives in reducing radiotherapy-related toxicity will also be explored. Methods Two datasets will be linked. (1) CHHiP (Conventional or Hypofractionated High Dose Intensity Modulated Radiotherapy for Prostate Cancer) randomised control trial. CHHiP contains pelvic symptoms and radiation-related toxicity reported by patients and clinicians. (2) GP (General Practice) data from RCGP RSC (Royal College of General Practitioners Research and Surveillance Centre). The GP records of CHHiP patients will be extracted, including cardiovascular co-morbidities, diabetes and prescription medications. Statistical analysis of the combined dataset will be performed in order to investigate the effect. Conclusions Linking two sources of healthcare data is an exciting area of big healthcare data research. With limited data in clinical trials (not all clinical trials collect information on co-morbidities or medications) and limited lengths of follow-up, linking different sources of information is increasingly needed to investigate long-term outcomes. With increasing pressures to collect detailed information in clinical trials (e.g. co-morbidities, medications), linkage to routinely collected data offers the potential to support efficient conduct of clinical trials.
Collapse
Key Words
- ANOVA, analysis of variance
- BNF, British National Formulary
- Big data
- CHHiP
- CHHiP, Conventional or Hypofractionated High Dose Intensity Modulated Radiotherapy for Prostate Cancer
- Data linkage
- EPIC, Expanded Prostate Cancer Index Composite
- FACT-P, Functional Assessment of Cancer Therapy-Prostate
- GEE, Generalized Estimating Equations
- GP, General Practitioner
- ICD10, International Classification of Disease version 10
- ICR, Institute of Cancer Research
- IMRT, Intensity Modulated Radiotherapy
- LENT/SOMA, Late Effects Normal Tissue Toxicity; subjective, objective, management, and analytic
- Late-effects
- PCa, Prostate cancer
- PROs, Patient Reported Outcomes
- QOL, Quality of life
- RCGP RSC
- RCGP, Royal College of General Practitioners
- RCT, Randomised Control Trial
- REC, Research Ethics Committee
- RSC, Research & Surveillance Centre
- RTOG, Radiation Therapy Oncology Group
- Radiotherapy-related side-effects
- SHA2-512, Secure Hash Algorithm 2 with 512 bit hash values
- UCLA-PCI, University of California, Los Angeles Prostate Cancer Index
- UK, United Kingdom
Collapse
Affiliation(s)
- Agnieszka Lemanska
- School of Health Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Rachel C Byford
- Department of Health Care Management and Policy, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Ana Correa
- Department of Health Care Management and Policy, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Clare Cruickshank
- The Institute of Cancer Research - Clinical Trials and Statistics Unit, London, UK
| | - David P Dearnaley
- The Institute of Cancer Research and Royal Marsden NHS Trust, London, UK
| | - Clare Griffin
- The Institute of Cancer Research - Clinical Trials and Statistics Unit, London, UK
| | - Emma Hall
- The Institute of Cancer Research - Clinical Trials and Statistics Unit, London, UK
| | - Simon de Lusignan
- Department of Health Care Management and Policy, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Sara Faithfull
- School of Health Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| |
Collapse
|
35
|
Diegeler S, Hellweg CE. Intercellular Communication of Tumor Cells and Immune Cells after Exposure to Different Ionizing Radiation Qualities. Front Immunol 2017. [PMID: 28638385 PMCID: PMC5461334 DOI: 10.3389/fimmu.2017.00664] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Ionizing radiation can affect the immune system in many ways. Depending on the situation, the whole body or parts of the body can be acutely or chronically exposed to different radiation qualities. In tumor radiotherapy, a fractionated exposure of the tumor (and surrounding tissues) is applied to kill the tumor cells. Currently, mostly photons, and also electrons, neutrons, protons, and heavier particles such as carbon ions, are used in radiotherapy. Tumor elimination can be supported by an effective immune response. In recent years, much progress has been achieved in the understanding of basic interactions between the irradiated tumor and the immune system. Here, direct and indirect effects of radiation on immune cells have to be considered. Lymphocytes for example are known to be highly radiosensitive. One important factor in indirect interactions is the radiation-induced bystander effect which can be initiated in unexposed cells by expression of cytokines of the irradiated cells and by direct exchange of molecules via gap junctions. In this review, we summarize the current knowledge about the indirect effects observed after exposure to different radiation qualities. The different immune cell populations important for the tumor immune response are natural killer cells, dendritic cells, and CD8+ cytotoxic T-cells. In vitro and in vivo studies have revealed the modulation of their functions due to ionizing radiation exposure of tumor cells. After radiation exposure, cytokines are produced by exposed tumor and immune cells and a modulated expression profile has also been observed in bystander immune cells. Release of damage-associated molecular patterns by irradiated tumor cells is another factor in immune activation. In conclusion, both immune-activating and -suppressing effects can occur. Enhancing or inhibiting these effects, respectively, could contribute to modified tumor cell killing after radiotherapy.
Collapse
Affiliation(s)
- Sebastian Diegeler
- Division of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), Köln, Germany
| | - Christine E Hellweg
- Division of Radiation Biology, Institute of Aerospace Medicine, German Aerospace Center (DLR), Köln, Germany
| |
Collapse
|
36
|
Kole AJ, Kole L, Moran MS. Acute radiation dermatitis in breast cancer patients: challenges and solutions. BREAST CANCER (DOVE MEDICAL PRESS) 2017; 9:313-323. [PMID: 28503074 PMCID: PMC5426474 DOI: 10.2147/bctt.s109763] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Nearly all women who receive radiotherapy (RT) for breast cancer experience some degree of radiation dermatitis. However, evidence describing the appropriate management of radiation dermatitis is often lacking or contradictory. Here, we summarize the available literature regarding radiation dermatitis causes, the presentation and timing of symptoms, methods for dermatitis assessment and prevention, and review evidence-based management strategies.
Collapse
Affiliation(s)
| | - Lauren Kole
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | | |
Collapse
|
37
|
Doi H, Matsumoto S, Odawara S, Shikata T, Kitajima K, Tanooka M, Takada Y, Tsujimura T, Kamikonya N, Hirota S. Pravastatin reduces radiation-induced damage in normal tissues. Exp Ther Med 2017; 13:1765-1772. [PMID: 28565765 PMCID: PMC5443166 DOI: 10.3892/etm.2017.4192] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 12/23/2016] [Indexed: 12/26/2022] Open
Abstract
Pravastatin is an inhibitor of 3-hydroxy-3-methyl- glutaryl-coenzyme A reductase that has been reported to have therapeutic applications in a range of inflammatory conditions. The aim of the present study was to assess the radioprotective effects of pravastatin in an experimental animal model. Mice were divided into two groups: The control group received ionizing radiation with no prior medication, while the pravastatin group received pravastatin prior to ionizing radiation. Pravastatin was administered orally at 30 mg/kg body weight in drinking water at 24 and 4 h before irradiation. Intestinal crypt epithelial cell survival and the incidence of apoptosis in the intestine and lung were measured post-irradiation. The effect of pravastatin on intestinal DNA damage was determined by immunohistochemistry. Finally, the effect of pravastatin on tumor response to radiotherapy was examined in a mouse mesothelioma xenograft model. Pravastatin increased the number of viable intestinal crypts and this effect was statistically significant in the ileum (P<0.0001). The pravastatin group showed significantly lower apoptotic indices in all examined parts of the intestine (P<0.0001) and tended to show reduced apoptosis in the lung. Pravastatin reduced the intestinal expression of ataxia-telangiectasia mutated and gamma-H2AX after irradiation. No apparent pravastatin-related differences were observed in the response of xenograft tumors to irradiation. In conclusion, pravastatin had radioprotective effects on the intestine and lung and reduced radiation-induced DNA double-strand breaks. Pravastatin may increase the therapeutic index of radiotherapy.
Collapse
Affiliation(s)
- Hiroshi Doi
- Department of Radiology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Seiji Matsumoto
- Department of Thoracic Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Soichi Odawara
- Department of Radiology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Toshiyuki Shikata
- Department of Pharmacy, Hyogo College of Medicine Sasayama Medical Center, Sasayama, Hyogo 669-2321, Japan
| | - Kazuhiro Kitajima
- Department of Radiology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Masao Tanooka
- Department of Radiological Technology, Hyogo College of Medicine College Hospital, Nishinomiya, Hyogo 663-8501, Japan
| | - Yasuhiro Takada
- Department of Radiology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Tohru Tsujimura
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Norihiko Kamikonya
- Department of Radiology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Shozo Hirota
- Department of Radiology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| |
Collapse
|
38
|
Current Status of Targeted Radioprotection and Radiation Injury Mitigation and Treatment Agents: A Critical Review of the Literature. Int J Radiat Oncol Biol Phys 2017; 98:662-682. [PMID: 28581409 DOI: 10.1016/j.ijrobp.2017.02.211] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/22/2017] [Accepted: 02/23/2017] [Indexed: 01/17/2023]
Abstract
As more cancer patients survive their disease, concerns about radiation therapy-induced side effects have increased. The concept of radioprotection and radiation injury mitigation and treatment offers the possibility to enhance the therapeutic ratio of radiation therapy by limiting radiation therapy-induced normal tissue injury without compromising its antitumor effect. Advances in the understanding of the underlying mechanisms of radiation toxicity have stimulated radiation oncologists to target these pathways across different organ systems. These generalized radiation injury mechanisms include production of free radicals such as superoxides, activation of inflammatory pathways, and vascular endothelial dysfunction leading to tissue hypoxia. There is a significant body of literature evaluating the effectiveness of various treatments in preventing, mitigating, or treating radiation-induced normal tissue injury. Whereas some reviews have focused on a specific disease site or agent, this critical review focuses on a mechanistic classification of activity and assesses multiple agents across different disease sites. The classification of agents used herein further offers a useful framework to organize the multitude of treatments that have been studied. Many commonly available treatments have demonstrated benefit in prevention, mitigation, and/or treatment of radiation toxicity and warrant further investigation. These drug-based approaches to radioprotection and radiation injury mitigation and treatment represent an important method of making radiation therapy safer.
Collapse
|
39
|
Appelbe OK, Zhang Q, Pelizzari CA, Weichselbaum RR, Kron SJ. Image-Guided Radiotherapy Targets Macromolecules through Altering the Tumor Microenvironment. Mol Pharm 2016; 13:3457-3467. [PMID: 27560921 DOI: 10.1021/acs.molpharmaceut.6b00465] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Current strategies to target tumors with nanomedicines rely on passive delivery via the enhanced permeability and retention effect, leveraging the disorganized tumor microvasculature to promote macromolecule extravasation and the reduced lymphatic and venous drainage that favor retention. Nonetheless, FDA approvals and clinical use of nanomedicines have lagged, reflecting failure to display superiority over conventional formulations. Here, we have exploited image-guided X-irradiation to augment nanoparticle accumulation in tumors. A single 5 Gy dose of radiation, below that required to significantly delay tumor growth, can markedly enhance delivery of macromolecules and nanoparticles. The radiation effect was independent of endothelial cell integrity, suggesting a primary role for damage to microvascular pericytes and/or interstitial extracellular matrix. Significantly, radiation-guided delivery potentiated the therapeutic effects of PEGylated liposomal doxorubicin on experimental tumors. Applied to patients, these results suggest repurposing image-guided radiotherapy as a tool to guide cancer nanomedicine delivery, enhancing local control for primary tumors and metastatic disease while limiting systemic toxicity.
Collapse
Affiliation(s)
- Oliver K Appelbe
- Ludwig Center for Metastasis Research, The University of Chicago , 5758 South Maryland Avenue, MC 9006, Chicago, Illinois 60637, United States.,Department of Molecular Genetics and Cellular Biology, The University of Chicago , 929 East 57th Street, GCIS W519, Chicago, Illinois 60637, United States
| | - Qingbei Zhang
- Ludwig Center for Metastasis Research, The University of Chicago , 5758 South Maryland Avenue, MC 9006, Chicago, Illinois 60637, United States.,Department of Molecular Genetics and Cellular Biology, The University of Chicago , 929 East 57th Street, GCIS W519, Chicago, Illinois 60637, United States
| | - Charles A Pelizzari
- Department of Radiation and Cellular Oncology, The University of Chicago , 5758 South Maryland Avenue, MC 9006, Chicago, Illinois 60637, United States
| | - Ralph R Weichselbaum
- Ludwig Center for Metastasis Research, The University of Chicago , 5758 South Maryland Avenue, MC 9006, Chicago, Illinois 60637, United States.,Department of Radiation and Cellular Oncology, The University of Chicago , 5758 South Maryland Avenue, MC 9006, Chicago, Illinois 60637, United States
| | - Stephen J Kron
- Ludwig Center for Metastasis Research, The University of Chicago , 5758 South Maryland Avenue, MC 9006, Chicago, Illinois 60637, United States.,Department of Molecular Genetics and Cellular Biology, The University of Chicago , 929 East 57th Street, GCIS W519, Chicago, Illinois 60637, United States
| |
Collapse
|
40
|
Protection against Radiotherapy-Induced Toxicity. Antioxidants (Basel) 2016; 5:antiox5030022. [PMID: 27399787 PMCID: PMC5039571 DOI: 10.3390/antiox5030022] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 06/27/2016] [Accepted: 06/28/2016] [Indexed: 01/18/2023] Open
Abstract
Radiation therapy is a highly utilized therapy in the treatment of malignancies with up to 60% of cancer patients receiving radiation therapy as a part of their treatment regimen. Radiation therapy does, however, cause a wide range of adverse effects that can be severe and cause permanent damage to the patient. In an attempt to minimize these effects, a small number of compounds have been identified and are in use clinically for the prevention and treatment of radiation associated toxicities. Furthermore, there are a number of emerging therapies being developed for use as agents that protect against radiation-induced toxicities. The aim of this review was to evaluate and summarise the evidence that exists for both the known radioprotectant agents and the agents that show promise as future radioprotectant agents.
Collapse
|
41
|
Morris KAL, Haboubi NY. Pelvic radiation therapy: Between delight and disaster. World J Gastrointest Surg 2015; 7:279-88. [PMID: 26649150 PMCID: PMC4663381 DOI: 10.4240/wjgs.v7.i11.279] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 08/10/2015] [Accepted: 10/01/2015] [Indexed: 02/07/2023] Open
Abstract
In the last few decades radiotherapy was established as one of the best and most widely used treatment modalities for certain tumours. Unfortunately that came with a price. As more people with cancer survive longer an ever increasing number of patients are living with the complications of radiotherapy and have become, in certain cases, difficult to manage. Pelvic radiation disease (PRD) can result from ionising radiation-induced damage to surrounding non-cancerous tissues resulting in disruption of normal physiological functions and symptoms such as diarrhoea, tenesmus, incontinence and rectal bleeding. The burden of PRD-related symptoms, which impact on a patient's quality of life, has been under appreciated and sub-optimally managed. This article serves to promote awareness of PRD and the vast potential there is to improve current service provision and research activities.
Collapse
|
42
|
Haubner F, Muschter D, Schuster N, Pohl F, Ahrens N, Prantl L, Gassner H. Platelet-rich plasma stimulates dermal microvascular endothelial cells and adipose derived stem cells after external radiation. Clin Hemorheol Microcirc 2015; 61:279-90. [DOI: 10.3233/ch-151982] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- F. Haubner
- Department of Otorhinolaryngology, Division of Facial Plastic Surgery, University of Regensburg, Germany
| | - D. Muschter
- Department of Otorhinolaryngology, Division of Facial Plastic Surgery, University of Regensburg, Germany
| | - N. Schuster
- Department of Otorhinolaryngology, Division of Facial Plastic Surgery, University of Regensburg, Germany
| | - F. Pohl
- Department of Radiotherapy, University of Regensburg, Germany
| | - N. Ahrens
- Department of Clinical Chemistry and Laboratory Medicine, University of Regensburg, Germany
| | - L. Prantl
- Center for Plastic, Aesthetic, Hand & Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - H.G. Gassner
- Department of Otorhinolaryngology, Division of Facial Plastic Surgery, University of Regensburg, Germany
| |
Collapse
|
43
|
Vascular Complications After Radiotherapy in Head and Neck Free Flap Reconstruction. Ann Plast Surg 2015; 75:309-15. [DOI: 10.1097/sap.0000000000000081] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
44
|
Lu P, Liu J, Pang X. Pravastatin inhibits fibrinogen- and FDP-induced inflammatory response via reducing the production of IL-6, TNF-α and iNOS in vascular smooth muscle cells. Mol Med Rep 2015; 12:6145-51. [PMID: 26238934 DOI: 10.3892/mmr.2015.4149] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 07/10/2015] [Indexed: 11/05/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory response of the arterial wall to pro‑atherosclerotic factors. As an inflammatory marker, fibrinogen directly participates in the pathogenesis of atherosclerosis. Our previous study demonstrated that fibrinogen and fibrin degradation products (FDP) produce a pro‑inflammatory effect on vascular smooth muscle cells (VSMCs) through inducing the production of interleukin‑6 (IL‑6), tumor necrosis factor‑α (TNF‑α) and inducible nitric oxide synthase (iNOS). In the present study, the effects of pravastatin on fibrinogen‑ and FDP‑induced expression of IL‑6, TNF‑α and iNOS were observed in VSMCs. The results showed that pravastatin dose‑dependently inhibited fibrinogen‑ and FDP‑stimulated expression of IL‑6, TNF‑α and iNOS in VSMCs at the mRNA and protein level. The maximal inhibition of protein expression of IL‑6, TNF‑α and iNOS was 46.9, 42.7 and 49.2% in fibrinogen‑stimulated VSMCs, and 50.2, 49.8 and 53.6% in FDP‑stimulated VSMCs, respectively. This suggests that pravastatin has the ability to relieve vascular inflammation via inhibiting the generation of IL‑6, TNF‑α and iNOS. The results of the present study may aid in further explaining the beneficial effects of pravastatin on atherosclerosis and related cardiovascular diseases. In addition, they suggest that application of pravastatin may be beneficial for prevention of atherosclerosis formation in hyperfibrinogenemia.
Collapse
Affiliation(s)
- Peipei Lu
- Department of Pharmacology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Juntian Liu
- Department of Pharmacology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Xiaoming Pang
- Department of Pharmacology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
45
|
Korpela E, Liu SK. Endothelial perturbations and therapeutic strategies in normal tissue radiation damage. Radiat Oncol 2014; 9:266. [PMID: 25518850 PMCID: PMC4279961 DOI: 10.1186/s13014-014-0266-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 11/18/2014] [Indexed: 02/08/2023] Open
Abstract
Most cancer patients are treated with radiotherapy, but the treatment can also damage the surrounding normal tissue. Radiotherapy side-effects diminish patients’ quality of life, yet effective biological interventions for normal tissue damage are lacking. Protecting microvascular endothelial cells from the effects of irradiation is emerging as a targeted damage-reduction strategy. We illustrate the concept of the microvasculature as a mediator of overall normal tissue radiation toxicity through cell death, vascular inflammation (hemodynamic and molecular changes) and a change in functional capacity. Endothelial cell targeted therapies that protect against such endothelial cell perturbations and the development of acute normal tissue damage are mostly under preclinical development. Since acute radiation toxicity is a common clinical problem in cutaneous, gastrointestinal and mucosal tissues, we also focus on damage in these tissues.
Collapse
Affiliation(s)
- Elina Korpela
- Biological Sciences, Sunnybrook Research Institute and Odette Cancer Centre, Sunnybrook Health Sciences Centre, 2075 Bayview Ave., Toronto, M4N 3M5, Canada. .,Department of Medical Biophysics, University of Toronto, 101 College St., Toronto, M5G 1L7, Canada.
| | - Stanley K Liu
- Biological Sciences, Sunnybrook Research Institute and Odette Cancer Centre, Sunnybrook Health Sciences Centre, 2075 Bayview Ave., Toronto, M4N 3M5, Canada. .,Department of Medical Biophysics, University of Toronto, 101 College St., Toronto, M5G 1L7, Canada. .,Department of Radiation Oncology, University of Toronto, 149 College St., Toronto, M5T 1P5, Canada.
| |
Collapse
|
46
|
Xu J, Cao Y. Radiation-induced carotid artery stenosis: a comprehensive review of the literature. INTERVENTIONAL NEUROLOGY 2014; 2:183-92. [PMID: 25337087 DOI: 10.1159/000363068] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In recent decades, with the improvement of radiotherapy (RT) technology and comprehensive treatment, the survival rate of head and neck malignancies has gained remarkable progress. Vascular injury and subsequent carotid stenosis following RT, as the backbone of treatment, have received increasing attention. Many investigations have demonstrated that radiation can result in the increase in carotid intima-media thickness, carotid stenosis and consequently lead to a higher risk of cerebrovascular events such as transient ischemic attack and stroke. In this review, we will examine the incidence of radiation-induced carotid artery stenosis, its morphological and histological characteristics, as well as its pathogenesis. The treatment and prevention methods, including follow-up strategies, will also be discussed at the end of the present review.
Collapse
Affiliation(s)
- Jiaping Xu
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China ; Institute of Neuroscience, Soochow University, Suzhou, China
| | - Yongjun Cao
- Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, China ; Institute of Neuroscience, Soochow University, Suzhou, China
| |
Collapse
|
47
|
Benderitter M, Caviggioli F, Chapel A, Coppes RP, Guha C, Klinger M, Malard O, Stewart F, Tamarat R, van Luijk P, Limoli CL. Stem cell therapies for the treatment of radiation-induced normal tissue side effects. Antioxid Redox Signal 2014; 21:338-55. [PMID: 24147585 PMCID: PMC4060814 DOI: 10.1089/ars.2013.5652] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Targeted irradiation is an effective cancer therapy but damage inflicted to normal tissues surrounding the tumor may cause severe complications. While certain pharmacologic strategies can temper the adverse effects of irradiation, stem cell therapies provide unique opportunities for restoring functionality to the irradiated tissue bed. RECENT ADVANCES Preclinical studies presented in this review provide encouraging proof of concept regarding the therapeutic potential of stem cells for treating the adverse side effects associated with radiotherapy in different organs. Early-stage clinical data for radiation-induced lung, bone, and skin complications are promising and highlight the importance of selecting the appropriate stem cell type to stimulate tissue regeneration. CRITICAL ISSUES While therapeutic efficacy has been demonstrated in a variety of animal models and human trials, a range of additional concerns regarding stem cell transplantation for ameliorating radiation-induced normal tissue sequelae remain. Safety issues regarding teratoma formation, disease progression, and genomic stability along with technical issues impacting disease targeting, immunorejection, and clinical scale-up are factors bearing on the eventual translation of stem cell therapies into routine clinical practice. FUTURE DIRECTIONS Follow-up studies will need to identify the best possible stem cell types for the treatment of early and late radiation-induced normal tissue injury. Additional work should seek to optimize cellular dosing regimes, identify the best routes of administration, elucidate optimal transplantation windows for introducing cells into more receptive host tissues, and improve immune tolerance for longer-term engrafted cell survival into the irradiated microenvironment.
Collapse
Affiliation(s)
- Marc Benderitter
- 1 Laboratory of Radiopathology and Experimental Therapies, IRSN , PRP-HOM, SRBE, Fontenay-aux-Roses, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Weigel C, Schmezer P, Plass C, Popanda O. Epigenetics in radiation-induced fibrosis. Oncogene 2014; 34:2145-55. [PMID: 24909163 DOI: 10.1038/onc.2014.145] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 04/17/2014] [Accepted: 04/23/2014] [Indexed: 02/06/2023]
Abstract
Radiotherapy is a major cancer treatment option but dose-limiting side effects such as late-onset fibrosis in the irradiated tissue severely impair quality of life in cancer survivors. Efforts to explain radiation-induced fibrosis, for example, by genetic variation remained largely inconclusive. Recently published molecular analyses on radiation response and fibrogenesis showed a prominent role of epigenetic gene regulation. This review summarizes the current knowledge on epigenetic modifications in fibrotic disease and radiation response, and it points out the important role for epigenetic mechanisms such as DNA methylation, microRNAs and histone modifications in the development of this disease. The synopsis illustrates the complexity of radiation-induced fibrosis and reveals the need for investigations to further unravel its molecular mechanisms. Importantly, epigenetic changes are long-term determinants of gene expression and can therefore support those mechanisms that induce and perpetuate fibrogenesis even in the absence of the initial damaging stimulus. Future work must comprise the interconnection of acute radiation response and long-lasting epigenetic effects in order to assess their role in late-onset radiation fibrosis. An improved understanding of the underlying biology is fundamental to better comprehend the origin of this disease and to improve both preventive and therapeutic strategies.
Collapse
Affiliation(s)
- C Weigel
- Department of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - P Schmezer
- Department of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - C Plass
- Department of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - O Popanda
- Department of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
49
|
Voswinkel J, Francois S, Simon JM, Benderitter M, Gorin NC, Mohty M, Fouillard L, Chapel A. Use of mesenchymal stem cells (MSC) in chronic inflammatory fistulizing and fibrotic diseases: a comprehensive review. Clin Rev Allergy Immunol 2014; 45:180-92. [PMID: 23296948 DOI: 10.1007/s12016-012-8347-6] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Mesenchymal stem cells (MSC), multipotent adult stem cells, feature the potential to regenerate tissue damage and, in parallel, inhibit inflammation and fibrosis. MSC can be safely transplanted in autologous and allogeneic ways as they are non-immunogenic, and consequently represent a therapeutic option for refractory connective tissue diseases, fibrosing diseases like scleroderma and fistulizing colitis like in Crohn's disease. Actually, there are more than 200 registered clinical trial sites for evaluating MSC therapy, and 22 are on autoimmune diseases. In irradiation-induced colitis, MSC accelerate functional recovery of the intestine and dampen the systemic inflammatory response. In order to provide rescue therapy for accidentally over-irradiated prostate cancer patients who underwent radiotherapy, allogeneic bone marrow-derived MSC from family donors were intravenously infused to three patients with refractory and fistulizing colitis resembling fistulizing Crohn's disease. Systemic MSC therapy of refractory irradiation-induced colitis was safe and effective on pain, diarrhoea, hemorrhage, inflammation and fistulization accompanied by modulation of the lymphocyte subsets towards an increase of T regulatory cells and a decrease of activated effector T cells. The current data indicate that MSC represent a promising alternative strategy in the treatment of various immune-mediated diseases. Encouraging results have already been obtained from clinical trials in Crohn's disease and SLE as well as from case series in systemic sclerosis. MSC represent a safe therapeutic measure for patients who suffer from chronic and fistulizing colitis. These findings are instructional for the management of refractory inflammatory bowel diseases that are characterized by similar clinical and immunopathological features.
Collapse
Affiliation(s)
- Jan Voswinkel
- Department of Hematology, Saint Antoine Hospital APHP and UPMC University, UMRS 938, 184 rue Faubourg Saint Antoine, 75012, Paris, France,
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Voswinkel J, Francois S, Gorin NC, Chapel A. Gastro-intestinal autoimmunity: preclinical experiences and successful therapy of fistulizing bowel diseases and gut Graft versus host disease by mesenchymal stromal cells. Immunol Res 2014; 56:241-8. [PMID: 23564182 DOI: 10.1007/s12026-013-8397-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mesenchymal stromal cells (MSC) are multipotent adult stem cells with the potential to regenerate tissue damage and inhibit inflammation and fibrosis in parallel. As they are non-immunogenic, MSC can be safely auto- and allotransplanted and consequently represent a therapeutic option for refractory connective tissue diseases and fistulizing colitis like Crohn's disease. Actually, there are more than 200 registered clinical trial sites for evaluating MSC therapy, 22 are on autoimmune diseases and 27 are actually recruiting bowel disease' patients. More than 1,500 patients with bowel diseases like Crohn's disease were treated in clinical trials by local as well as systemic MSC therapy. Phase I and II trials on fistula documented the feasibility and safety of MSC therapy, and a significant superiority compared to fibrin glue in fistulizing bowel diseases was demonstrated. Autologous as well as allogeneic use of Bone marrow as well as of adipose tissue-derived MSC are feasible. In refractory Graft versus host disease, especially in refractory gut Graft versus host diseases, encouraging results were reported using MSC. Systemic MSC therapy of refractory irradiation-induced colitis was safe and effective on pain, diarrhea, hemorrhage, inflammation and fistulization accompanied by modulation of the lymphocyte subsets toward an increase in T regulatory cells and a decrease in activated effector T cells. Mesenchymal stem cells represent a safe therapy for patients with refractory inflammatory bowel diseases.
Collapse
Affiliation(s)
- Jan Voswinkel
- Department of Haematology, Saint Antoine Hospital, APHP and UPMC University, UMRS 938, 184 rue Faubourg Saint Antoine, 75012, Paris, France.
| | | | | | | |
Collapse
|