1
|
Hardy SJ, Finkelstein A, Milano MT, Schifitto G, Sun H, Holley K, Usuki K, Weber MT, Zheng D, Seplaki CL, Janelsins M. Association of Radiation Dose to the Amygdala-Orbitofrontal Network with Emotion Recognition Task Performance in Patients with Low-Grade and Benign Brain Tumors. Cancers (Basel) 2023; 15:5544. [PMID: 38067248 PMCID: PMC10705220 DOI: 10.3390/cancers15235544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/16/2023] [Accepted: 11/18/2023] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Although data are limited, difficulty in social cognition occurs in up to 83% of patients with brain tumors. It is unknown whether cranial radiation therapy (RT) dose to the amygdala-orbitofrontal network can impact social cognition. METHODS We prospectively enrolled 51 patients with low-grade and benign brain tumors planned for cranial RT. We assessed longitudinal changes on an emotion recognition task (ERT) that measures the ability to recognize emotional states by displaying faces expressing six basic emotions and their association with the RT dose to the amygdala-orbitofrontal network. ERT outcomes included the median time to choose a response (ERTOMDRT) or correct response (ERTOMDCRT) and total correct responses (ERTHH). RESULTS The RT dose to the amygdala-orbitofrontal network was significantly associated with longer median response times on the ERT. Increases in median response times occurred at lower doses than decreases in total correct responses. The medial orbitofrontal cortex was the most important variable on regression trees predicting change in the ERTOMDCRT. DISCUSSION This is, to our knowledge, the first study to show that off-target RT dose to the amygdala-orbitofrontal network is associated with performance on a social cognition task, a facet of cognition that has previously not been mechanistically studied after cranial RT.
Collapse
Affiliation(s)
- Sara J. Hardy
- Department of Radiation Oncology, University of Rochester, Rochester, NY 14620, USA; (M.T.M.); (D.Z.); (M.J.)
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - Alan Finkelstein
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, USA;
- Center for Advanced Brain Imaging and Neurophysiology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Michael T. Milano
- Department of Radiation Oncology, University of Rochester, Rochester, NY 14620, USA; (M.T.M.); (D.Z.); (M.J.)
| | - Giovanni Schifitto
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - Hongying Sun
- Department of Surgery, Supportive Care in Cancer, University of Rochester Medical Center, Rochester, NY 14642, USA; (H.S.); (M.T.W.)
| | - Koren Holley
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - Kenneth Usuki
- Department of Radiation Oncology, University of Rochester, Rochester, NY 14620, USA; (M.T.M.); (D.Z.); (M.J.)
| | - Miriam T. Weber
- Department of Surgery, Supportive Care in Cancer, University of Rochester Medical Center, Rochester, NY 14642, USA; (H.S.); (M.T.W.)
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Dandan Zheng
- Department of Radiation Oncology, University of Rochester, Rochester, NY 14620, USA; (M.T.M.); (D.Z.); (M.J.)
| | - Christopher L. Seplaki
- Department of Public Health Sciences, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Office for Aging Research and Health Services, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Michelle Janelsins
- Department of Radiation Oncology, University of Rochester, Rochester, NY 14620, USA; (M.T.M.); (D.Z.); (M.J.)
- Department of Surgery, Supportive Care in Cancer, University of Rochester Medical Center, Rochester, NY 14642, USA; (H.S.); (M.T.W.)
| |
Collapse
|
2
|
Li Y, Zhou Z, Xu S, Jiang J, Xiao J. Review of the Pathogenesis, Diagnosis, and Management of Osteoradionecrosis of the Femoral Head. Med Sci Monit 2023; 29:e940264. [PMID: 37310931 PMCID: PMC10276533 DOI: 10.12659/msm.940264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 04/13/2023] [Indexed: 06/15/2023] Open
Abstract
Osteoradionecrosis (ORN) of the femoral head is an important issue for orthopedists and radiologists in clinical practice. With the rapid development of technological advances in radiation therapy and the improvement in cancer survival rates, the incidence of ORN is rising, and there is an unmet need for basic and clinical research. The pathogenesis of ORN is complex, and includes vascular injury, mesenchymal stem cell injury, bone loss, reactive oxygen species, radiation-induced fibrosis, and cell senescence. The diagnosis of ORN is challenging and requires multiple considerations, including exposure to ionizing radiation, clinical manifestations, and findings on physical examination and imaging. Differential diagnosis is essential, as clinical symptoms of ORN of the femoral head can resemble many other hip conditions. Hyperbaric oxygen therapy, total hip arthroplasty, and Girdlestone resection arthroplasty are effective treatments, each with their own advantages and disadvantages. The literature on ORN of the femoral head is incomplete and there is no criterion standard or clear consensus on management. Clinicians should gain a better and more comprehensive understanding on this disease to facilitate its early and better prevention, diagnosis, and treatment. This article aims to review the pathogenesis, diagnosis, and management of osteoradionecrosis of the femoral head.
Collapse
Affiliation(s)
- Yang Li
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, PR China
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, PR China
| | - Zhongsheng Zhou
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, PR China
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, PR China
| | - Shenghao Xu
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, PR China
| | - Jinlan Jiang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin, PR China
| | - Jianlin Xiao
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, PR China
| |
Collapse
|
3
|
Choi YY, Kim A, Lee Y, Lee YH, Park M, Shin E, Park S, Youn B, Seong KM. The miR-126-5p and miR-212-3p in the extracellular vesicles activate monocytes in the early stage of radiation-induced vascular inflammation implicated in atherosclerosis. J Extracell Vesicles 2023; 12:e12325. [PMID: 37140946 PMCID: PMC10158827 DOI: 10.1002/jev2.12325] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 04/04/2023] [Accepted: 04/12/2023] [Indexed: 05/05/2023] Open
Abstract
People exposed to radiation in cancer therapy and nuclear accidents are at increased risk of cardiovascular outcomes in long-term survivors. Extracellular vesicles (EVs) are involved in radiation-induced endothelial dysfunction, but their role in the early stage of vascular inflammation after radiation exposure remains to be fully understood. Herein, we demonstrate that endothelial cell-derived EVs containing miRNAs initiate monocyte activation in radiation-induced vascular inflammation. In vitro co-culture and in vivo experimental data showed that endothelial EVs can be sensitively increased by radiation exposure in a dose-dependent manner, and stimulate monocytes releasing monocytic EVs and adhesion to endothelial cells together with an increase in the expression of genes encoding specific ligands for cell-cell interaction. Small RNA sequencing and transfection using mimics and inhibitors explained that miR-126-5p and miR-212-3p enriched in endothelial EVs initiate vascular inflammation by monocyte activation after radiation exposure. Moreover, miR-126-5p could be detected in the circulating endothelial EVs of radiation-induced atherosclerosis model mice, which was found to be tightly correlated with the atherogenic index of plasma. In summary, our study showed that miR-126-5p and miR-212-3p present in the endothelial EVs mediate the inflammatory signals to activate monocytes in radiation-induced vascular injury. A better understanding of the circulating endothelial EVs content can promote their use as diagnostic and prognostic biomarkers for atherosclerosis after radiation exposure.
Collapse
Affiliation(s)
- You Yeon Choi
- Laboratory of Biological Dosimetry, National Radiation Emergency Medical Center, KIRAMS, Seoul, Republic of Korea
| | - Areumnuri Kim
- Laboratory of Radiation Exposure and Therapeutics, National Radiation Emergency Medical Center, KIRAMS, Seoul, Republic of Korea
| | - Younghyun Lee
- Laboratory of Biological Dosimetry, National Radiation Emergency Medical Center, KIRAMS, Seoul, Republic of Korea
| | - Yang Hee Lee
- Laboratory of Biological Dosimetry, National Radiation Emergency Medical Center, KIRAMS, Seoul, Republic of Korea
| | - Mineon Park
- Laboratory of Biological Dosimetry, National Radiation Emergency Medical Center, KIRAMS, Seoul, Republic of Korea
| | - Eunguk Shin
- Department of Biological Sciences, Pusan National University, Busan, Republic of Korea
| | - Sunhoo Park
- Laboratory of Biological Dosimetry, National Radiation Emergency Medical Center, KIRAMS, Seoul, Republic of Korea
| | - BuHyun Youn
- Department of Biological Sciences, Pusan National University, Busan, Republic of Korea
| | - Ki Moon Seong
- Laboratory of Biological Dosimetry, National Radiation Emergency Medical Center, KIRAMS, Seoul, Republic of Korea
| |
Collapse
|
4
|
Hardy SJ, Finkelstein AJ, Tivarus M, Culakova E, Mohile N, Weber M, Lin E, Zhong J, Usuki K, Schifitto G, Milano M, Janelsins-Benton MC. Cognitive and neuroimaging outcomes in individuals with benign and low-grade brain tumours receiving radiotherapy: a protocol for a prospective cohort study. BMJ Open 2023; 13:e066458. [PMID: 36792323 PMCID: PMC9933762 DOI: 10.1136/bmjopen-2022-066458] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 01/27/2023] [Indexed: 02/17/2023] Open
Abstract
INTRODUCTION Radiation-induced cognitive decline (RICD) occurs in 50%-90% of adult patients 6 months post-treatment. In patients with low-grade and benign tumours with long expected survival, this is of paramount importance. Despite advances in radiation therapy (RT) treatment delivery, better understanding of structures important for RICD is necessary to improve cognitive outcomes. We hypothesise that RT may affect network topology and microstructural integrity on MRI prior to any gross anatomical or apparent cognitive changes. In this longitudinal cohort study, we aim to determine the effects of RT on brain structural and functional integrity and cognition. METHODS AND ANALYSIS This study will enroll patients with benign and low-grade brain tumours receiving partial brain radiotherapy. Patients will receive either hypofractionated (>2 Gy/fraction) or conventionally fractionated (1.8-2 Gy/fraction) RT. All participants will be followed for 12 months, with MRIs conducted pre-RT and 6-month and 12 month post-RT, along with a battery of neurocognitive tests and questionnaires. The study was initiated in late 2018 and will continue enrolling through 2024 with final follow-ups completing in 2025. The neurocognitive battery assesses visual and verbal memory, attention, executive function, processing speed and emotional cognition. MRI protocols incorporate diffusion tensor imaging and resting state fMRI to assess structural connectivity and functional connectivity, respectively. We will estimate the association between radiation dose, imaging metrics and cognitive outcomes. ETHICS AND DISSEMINATION This study has been approved by the Research Subjects Review Board at the University of Rochester (STUDY00001512: Cognitive changes in patients receiving partial brain radiation). All results will be published in peer-reviewed journals and at scientific conferences. TRIAL REGISTRATION NUMBER ClinicalTrials.gov NCT04390906.
Collapse
Affiliation(s)
- Sara J Hardy
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York, USA
- Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA
| | - Alan J Finkelstein
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA
- Center for Advanced Brain Imaging and Neurophysiology, University of Rochester Medical Center, Rochester, New York, USA
| | - Madalina Tivarus
- Center for Advanced Brain Imaging and Neurophysiology, University of Rochester Medical Center, Rochester, New York, USA
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, New York, USA
| | - Eva Culakova
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Nimish Mohile
- Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA
| | - Miriam Weber
- Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, New York, USA
| | - Edward Lin
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, New York, USA
| | - Jianhui Zhong
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA
- Center for Advanced Brain Imaging and Neurophysiology, University of Rochester Medical Center, Rochester, New York, USA
| | - Kenneth Usuki
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York, USA
| | - Giovanni Schifitto
- Department of Neurology, Department of Imaging Sciences, University of Rochester Medical Center, Rochester, New York, USA
| | - Michael Milano
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York, USA
| | - M C Janelsins-Benton
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York, USA
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
5
|
Patient Derived Ex-Vivo Cancer Models in Drug Development, Personalized Medicine, and Radiotherapy. Cancers (Basel) 2022; 14:cancers14123006. [PMID: 35740672 PMCID: PMC9220792 DOI: 10.3390/cancers14123006] [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: 05/20/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary This review article highlights gaps in the current system of drug development and personalized medicine for cancer therapy. The ex vivo model system using tissue biopsy from patients will advance the development of the predictive disease specific biomarker, drug screening and assessment of treatment response on a personalized basis. Although this ex vivo system demonstrated promises, there are challenges and limitations which need to be mitigated for further advancement and better applications. Abstract The field of cancer research is famous for its incremental steps in improving therapy. The consistent but slow rate of improvement is greatly due to its meticulous use of consistent cancer biology models. However, as we enter an era of increasingly personalized cancer care, including chemo and radiotherapy, our cancer models must be equally able to be applied to all individuals. Patient-derived organoid (PDO) and organ-in-chip (OIC) models based on the micro-physiological bioengineered platform have already been considered key components for preclinical and translational studies. Accounting for patient variability is one of the greatest challenges in the crossover from preclinical development to clinical trials and patient derived organoids may offer a steppingstone between the two. In this review, we highlight how incorporating PDO’s and OIC’s into the development of cancer therapy promises to increase the efficiency of our therapeutics.
Collapse
|
6
|
Luft T, Dreger P, Radujkovic A. Endothelial cell dysfunction: a key determinant for the outcome of allogeneic stem cell transplantation. Bone Marrow Transplant 2021; 56:2326-2335. [PMID: 34253879 PMCID: PMC8273852 DOI: 10.1038/s41409-021-01390-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 06/08/2021] [Accepted: 06/22/2021] [Indexed: 01/26/2023]
Abstract
Allogeneic hematopoietic stem cell transplantation (alloSCT) carries the promise of cure for many malignant and non-malignant diseases of the lympho-hematopoietic system. Although outcome has improved considerably since the pioneering Seattle achievements more than 5 decades ago, non-relapse mortality (NRM) remains a major burden of alloSCT. There is increasing evidence that endothelial dysfunction is involved in many of the life-threatening complications of alloSCT, such as sinusoidal obstruction syndrome/venoocclusive disease, transplant-associated thrombotic microangiopathy, and refractory acute graft-versus host disease. This review delineates the role of the endothelium in severe complications after alloSCT and describes the current status of search for biomarkers predicting endothelial complications, including markers of endothelial vulnerability and markers of endothelial injury. Finally, implications of our current understanding of transplant-associated endothelial pathology for prevention and management of complications after alloSCT are discussed.
Collapse
Affiliation(s)
- Thomas Luft
- Department Medicine V, University of Heidelberg, Heidelberg, Germany.
| | - Peter Dreger
- Department Medicine V, University of Heidelberg, Heidelberg, Germany.
| | | |
Collapse
|
7
|
Sun X, Feinberg MW. Vascular Endothelial Senescence: Pathobiological Insights, Emerging Long Noncoding RNA Targets, Challenges and Therapeutic Opportunities. Front Physiol 2021; 12:693067. [PMID: 34220553 PMCID: PMC8242592 DOI: 10.3389/fphys.2021.693067] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/07/2021] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is a stable form of cell cycle arrest in response to various stressors. While it serves as an endogenous pro-resolving mechanism, detrimental effects ensue when it is dysregulated. In this review, we introduce recent advances for cellular senescence and inflammaging, the underlying mechanisms for the reduction of nicotinamide adenine dinucleotide in tissues during aging, new knowledge learned from p16 reporter mice, and the development of machine learning algorithms in cellular senescence. We focus on pathobiological insights underlying cellular senescence of the vascular endothelium, a critical interface between blood and all tissues. Common causes and hallmarks of endothelial senescence are highlighted as well as recent advances in endothelial senescence. The regulation of cellular senescence involves multiple mechanistic layers involving chromatin, DNA, RNA, and protein levels. New targets are discussed including the roles of long noncoding RNAs in regulating endothelial cellular senescence. Emerging small molecules are highlighted that have anti-aging or anti-senescence effects in age-related diseases and impact homeostatic control of the vascular endothelium. Lastly, challenges and future directions are discussed including heterogeneity of endothelial cells and endothelial senescence, senescent markers and detection of senescent endothelial cells, evolutionary differences for immune surveillance in mice and humans, and long noncoding RNAs as therapeutic targets in attenuating cellular senescence. Accumulating studies indicate that cellular senescence is reversible. A better understanding of endothelial cellular senescence through lifestyle and pharmacological interventions holds promise to foster a new frontier in the management of cardiovascular disease risk.
Collapse
Affiliation(s)
- Xinghui Sun
- Department of Biochemistry, University of Nebraska–Lincoln, Lincoln, NE, United States
- Nebraska Center for the Prevention of Obesity Diseases Through Dietary Molecules, University of Nebraska–Lincoln, Lincoln, NE, United States
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska–Lincoln, Lincoln, NE, United States
| | - Mark W. Feinberg
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
8
|
Xiong W, Liao Y, Qin JY, Li WH, Tang ZY. Adverse effects of chemoradiotherapy on invasion and metastasis of tumor cells. Genes Dis 2020; 7:351-358. [PMID: 32884989 PMCID: PMC7452502 DOI: 10.1016/j.gendis.2020.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 03/18/2020] [Accepted: 04/03/2020] [Indexed: 12/21/2022] Open
Abstract
The phenomenon of enhanced invasion and metastasis of residual tumor cells has been observed in an increasing number of patients receiving chemoradiotherapy recently, and tumor metastasis will undoubtedly limit patient prognosis. However, the key mechanism by which chemoradiotherapy affects the invasion and metastasis of tumor cells remains unclear. Studies have shown that chemoradiotherapy may directly act on tumor cells and alter the tumor microenvironment, or induce cell apoptosis and autophagy to promote tumor cell survival and metastasis. In this review, we summarize the potential mechanisms by which chemoradiotherapy may affect the biological behavior of tumor cells and open up new avenues for reducing tumor recurrence and metastasis after treatment. These insights will improve the efficacy of chemoradiotherapy.
Collapse
Affiliation(s)
- Wei Xiong
- The Department of Radiation Oncology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yong Liao
- Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ji-Yong Qin
- The Department of Radiation Oncology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wen-Hui Li
- The Department of Radiation Oncology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhao-You Tang
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Venkatesulu BP, Sanders KL, Hsieh C, Kim BK, Krishnan S. Biomarkers of radiation-induced vascular injury. Cancer Rep (Hoboken) 2019; 2:e1152. [PMID: 32721134 PMCID: PMC7941417 DOI: 10.1002/cnr2.1152] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 10/31/2018] [Accepted: 10/31/2018] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Cancer survivorship has thrown the spotlight on the incidence of nonmalignant chronic diseases in cancer patients. Endothelial injury is increasingly recognized as a consequence of cancer treatment, particularly after radiation therapy (RT). This review is to provide a current understanding on the pathophysiological mechanisms and predictive biomarkers of radiation-induced vascular injury. RECENT FINDINGS Radiation directly impacts vasculature by causing endothelial apoptosis and senescence, and alterations in normal homeostasis. This altered milieu at the endothelial surface may contribute to a systemic chronic inflammatory state that is superimposed upon the cascade of normal senescence processes leading to acceleration of age-related disorders, atherosclerosis, and chronic fibrosis. Vasculature imaging, blood-based or cell-component biomarkers, and signatures of genomics, proteomics, metabolomics, and radiomics are potential tools for detection of vascular damage after irradiation. CONCLUSIONS Development of a valid prediction model by combining an array of imaging tools, blood-based biomarkers, coupled with novel predictors like exosomes and metabolic degradation products can serve to identify RT-induced vascular injury early for subsequent introduction of newer therapeutic approaches to counter radiation morbidity.
Collapse
Affiliation(s)
- Bhanu Prasad Venkatesulu
- Departments of Experimental Radiation OncologyUniversity of Texas MD Anderson Cancer CenterHoustonTexas
| | - Keith L. Sanders
- Departments of Experimental Radiation OncologyUniversity of Texas MD Anderson Cancer CenterHoustonTexas
| | - Cheng‐En Hsieh
- Departments of Experimental Radiation OncologyUniversity of Texas MD Anderson Cancer CenterHoustonTexas
- Radiation OncologyUniversity of Texas MD Anderson Cancer CenterHoustonTexas
- The University of Texas MD Anderson Cancer Center‐UT Health Graduate School of Biomedical SciencesHoustonTexas
- Departments of Radiation Oncology, Chang Gung Memorial HospitalLinkou and Chang Gung UniversityTaoyuanTaiwan, ROC
| | - Byung Kyu Kim
- Departments of Experimental Radiation OncologyUniversity of Texas MD Anderson Cancer CenterHoustonTexas
- The University of Texas MD Anderson Cancer Center‐UT Health Graduate School of Biomedical SciencesHoustonTexas
| | - Sunil Krishnan
- Departments of Experimental Radiation OncologyUniversity of Texas MD Anderson Cancer CenterHoustonTexas
- Radiation OncologyUniversity of Texas MD Anderson Cancer CenterHoustonTexas
- The University of Texas MD Anderson Cancer Center‐UT Health Graduate School of Biomedical SciencesHoustonTexas
| |
Collapse
|