201
|
Morrison MA, Mueller S, Felton E, Jakary A, Stoller S, Avadiappan S, Yuan J, Molinaro AM, Braunstein S, Banerjee A, Hess CP, Lupo JM. Rate of radiation-induced microbleed formation on 7T MRI relates to cognitive impairment in young patients treated with radiation therapy for a brain tumor. Radiother Oncol 2020; 154:145-153. [PMID: 32966846 DOI: 10.1016/j.radonc.2020.09.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/04/2020] [Accepted: 09/14/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Radiation therapy (RT) is essential to the management of many brain tumors, but has been known to lead to cognitive decline and vascular injury in the form of cerebral microbleeds (CMBs). PURPOSE In a subset of children, adolescents, and young adults recruited from a larger trial investigating arteriopathy and stroke risk after RT, we evaluated the prevalence of CMBs after RT, examined risk factors for CMBs and cognitive impairment, and related their longitudinal development to cognitive performance changes. METHODS Twenty-five patients (mean 17 years, range: 10-25 years) underwent 7-Tesla MRI and cognitive assessment. Nineteen patients were treated with whole-brain or focal RT 1-month to 20-years prior, while 6 non-irradiated patients with posterior-fossa tumors served as controls. CMBs were detected on 7T susceptibility-weighted imaging (SWI) using semi-automated software, a first use in this population. RESULTS CMB detection sensitivity with 7T SWI was higher than previously reported at lower field strengths, with one or more CMBs detected in 100% of patients treated with RT at least 1-year prior. CMBs were localized to dose-targeted brain volumes with risk factors including whole-brain RT (p = 0.05), a higher RT dose (p = 0.01), increasing time since RT (p = 0.03), and younger age during RT (p = 0.01). Apart from RT dose, these factors were associated with impaired memory performance. Follow-up data in a subset of patients revealed a proportional increase in CMB count with worsening verbal memory performance (r = -0.85, p = 0.03). CONCLUSIONS Treatment with RT during youth is associated with the chronic development of CMBs that evolve with memory impairment over time.
Collapse
Affiliation(s)
- Melanie A Morrison
- Department of Radiology and Biomedical Imaging, University of California San Francisco, USA
| | - Sabine Mueller
- Department of Neurology, University of California San Francisco, USA
| | - Erin Felton
- Department of Neurology, University of California San Francisco, USA
| | - Angela Jakary
- Department of Radiology and Biomedical Imaging, University of California San Francisco, USA
| | - Schuyler Stoller
- Department of Neurology, University of California San Francisco, USA
| | - Sivakami Avadiappan
- Department of Radiology and Biomedical Imaging, University of California San Francisco, USA
| | - Justin Yuan
- Department of Radiology and Biomedical Imaging, University of California San Francisco, USA
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California San Francisco, USA; Department of Epidemiology & Biostatistics, University of California San Francisco, USA
| | - Steve Braunstein
- Department of Radiation Oncology, University of California San Francisco, USA
| | - Anu Banerjee
- Department of Neurology, University of California San Francisco, USA
| | - Christopher P Hess
- Department of Radiology and Biomedical Imaging, University of California San Francisco, USA; Department of Neurology, University of California San Francisco, USA
| | - Janine M Lupo
- Department of Radiology and Biomedical Imaging, University of California San Francisco, USA.
| |
Collapse
|
202
|
Parihar VK, Angulo MC, Allen BD, Syage A, Usmani MT, Passerat de la Chapelle E, Amin AN, Flores L, Lin X, Giedzinski E, Limoli CL. Sex-Specific Cognitive Deficits Following Space Radiation Exposure. Front Behav Neurosci 2020; 14:535885. [PMID: 33192361 PMCID: PMC7525092 DOI: 10.3389/fnbeh.2020.535885] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 08/24/2020] [Indexed: 12/17/2022] Open
Abstract
The radiation fields in space define tangible risks to the health of astronauts, and significant work in rodent models has clearly shown a variety of exposure paradigms to compromise central nervous system (CNS) functionality. Despite our current knowledge, sex differences regarding the risks of space radiation exposure on cognitive function remain poorly understood, which is potentially problematic given that 30% of astronauts are women. While work from us and others have demonstrated pronounced cognitive decrements in male mice exposed to charged particle irradiation, here we show that female mice exhibit significant resistance to adverse neurocognitive effects of space radiation. The present findings indicate that male mice exposed to low doses (≤30 cGy) of energetic (400 MeV/n) helium ions (4He) show significantly higher levels of neuroinflammation and more extensive cognitive deficits than females. Twelve weeks following 4He ion exposure, irradiated male mice demonstrated significant deficits in object and place recognition memory accompanied by activation of microglia, marked upregulation of hippocampal Toll-like receptor 4 (TLR4), and increased expression of the pro-inflammatory marker high mobility group box 1 protein (HMGB1). Additionally, we determined that exposure to 4He ions caused a significant decline in the number of dendritic branch points and total dendritic length along with the hippocampus neurons in female mice. Interestingly, only male mice showed a significant decline of dendritic spine density following irradiation. These data indicate that fundamental differences in inflammatory cascades between male and female mice may drive divergent CNS radiation responses that differentially impact the structural plasticity of neurons and neurocognitive outcomes following cosmic radiation exposure.
Collapse
Affiliation(s)
- Vipan K Parihar
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Maria C Angulo
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Barrett D Allen
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Amber Syage
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Manal T Usmani
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | | | - Amal Nayan Amin
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Lidia Flores
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Xiaomeng Lin
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Erich Giedzinski
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
203
|
Baker S, Logie N, Paulson K, Duimering A, Murtha A. Radiotherapy for Brain Tumors: Current Practice and Future Directions. CURRENT CANCER THERAPY REVIEWS 2020. [DOI: 10.2174/1573394715666181129105542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Radiotherapy is an important component of the treatment for primary and metastatic
brain tumors. Due to the close proximity of critical structures and normal brain parenchyma, Central
Nervous System (CNS) radiotherapy is associated with adverse effects such as neurocognitive
deficits, which must be weighed against the benefit of improved tumor control. Advanced radiotherapy
technology may help to mitigate toxicity risks, although there is a paucity of high-level
evidence to support its use. Recent advances have been made in the treatment for gliomas, meningiomas,
benign tumors, and metastases, although outcomes remain poor for many high grade
tumors. This review highlights recent developments in CNS radiotherapy, discusses common
treatment toxicities, critically reviews advanced radiotherapy technologies, and highlights promising
treatment strategies to improve clinical outcomes in the future.
Collapse
Affiliation(s)
- Sarah Baker
- Department of Radiation Oncology, Cross Cancer Institute, Edmonton, AB, Canada
| | - Natalie Logie
- University of Florida Proton Therapy Institute, Jacksonville, FL, United States
| | - Kim Paulson
- Department of Radiation Oncology, Cross Cancer Institute, Edmonton, AB, Canada
| | - Adele Duimering
- Department of Radiation Oncology, Cross Cancer Institute, Edmonton, AB, Canada
| | - Albert Murtha
- Department of Radiation Oncology, Cross Cancer Institute, Edmonton, AB, Canada
| |
Collapse
|
204
|
The acceleration of ageing in older patients with cancer. J Geriatr Oncol 2020; 12:343-351. [PMID: 32933870 DOI: 10.1016/j.jgo.2020.09.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/04/2020] [Accepted: 09/05/2020] [Indexed: 01/29/2023]
Abstract
Older cancer survivors may experience ageing at an accelerated rate when compared to their similar-aged, cancer-free counterparts. Ageing is undoubtedly a significant risk factor for cancer. There is evidence to suggest, however, that this relationship may in fact be bidirectional, with cancer and its treatments contributing to the ageing process. In this review, we outline the current literature linking cancer and anti-cancer therapy to adverse ageing outcomes and explore what additional research is needed in order to conclusively define cancer and its treatment as an accelerator of ageing.
Collapse
|
205
|
Emili M, Guidi S, Uguagliati B, Giacomini A, Bartesaghi R, Stagni F. Treatment with the flavonoid 7,8-Dihydroxyflavone: a promising strategy for a constellation of body and brain disorders. Crit Rev Food Sci Nutr 2020; 62:13-50. [DOI: 10.1080/10408398.2020.1810625] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Marco Emili
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Sandra Guidi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Beatrice Uguagliati
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Andrea Giacomini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Renata Bartesaghi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Fiorenza Stagni
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| |
Collapse
|
206
|
Huang X, Zhang X, Wang X, Rong X, Li Y, Li H, Jiang J, Cai J, Zhuo X, Pi Y, Lin J, Chua MLK, Argyriou AA, Lattanzi S, Simone CB, Glass J, Palmer JD, Chow E, Brown PD, Yue Z, Tang Y. A nomogram to predict symptomatic epilepsy in patients with radiation-induced brain necrosis. Neurology 2020; 95:e1392-e1403. [PMID: 32631922 DOI: 10.1212/wnl.0000000000010190] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 03/11/2020] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE To develop and validate a nomogram to predict epilepsy in patients with radiation-induced brain necrosis (RN). METHODS The nomogram was based on a retrospective analysis of 302 patients who were diagnosed with symptomatic RN from January 2005 to January 2016 in Sun Yat-sen Memorial Hospital using the Cox proportional hazards model. Discrimination of the nomogram was assessed by the concordance index (C index) and the calibration curve. The results were internally validated using bootstrap resampling and externally validated using 128 patients with RN from 2 additional hospitals. RESULTS A total of 302 patients with RN with a median follow-up of 3.43 years (interquartile range 2.54-5.45) were included in the training cohort; 65 (21.5%) developed symptomatic epilepsy during follow-up. Seven variables remained significant predictors of epilepsy after multivariable analyses: MRI lesion volume, creatine phosphokinase, the maximum radiation dose to the temporal lobe, RN treatment, history of hypertension and/or diabetes, sex, and total cholesterol level. In the validation cohort, 28 out of 128 (21.9%) patients had epilepsy after RN within a median follow-up of 3.2 years. The nomogram showed comparable discrimination between the training and validation cohort (corrected C index 0.76 [training] vs 0.72 [95% confidence interval 0.62-0.81; validation]). CONCLUSION Our study developed an easily applied nomogram for the prediction of RN-related epilepsy in a large RN cohort. CLASSIFICATION OF EVIDENCE This study provides Class III evidence that a nomogram predicts post-RN epilepsy.
Collapse
Affiliation(s)
- Xiaolong Huang
- From the Department of Neurology Bioland Laboratory (X.H., X. Zhang, X.R., Y.L., H.L., J.J., J.C., X. Zhuo, X.P., J.L., Z.Y., Y.T.) and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center (Y.T.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University; Guangdong Province Key Laboratory of Brain Function and Disease (Y.T.), Zhongshan School of Medicine, Sun Yat-Sen University; Department of Oncology (X.W.), The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Division of Radiation Oncology and Medical Sciences (M.L.K.C.), National Cancer Centre Singapore; Oncology Academic Programme (M.L.K.C.), Duke-NUS Medical School, Singapore; Department of Neurology (A.A.A.), Saint Andrew's State General Hospital of Patras, Greece; Neurological Clinic, Department of Experimental and Clinical Medicine (S.L.), Marche Polytechnic University, Italy; New York Proton Center (C.B.S.), New York; Thomas Jefferson University (J.G.), Philadelphia, PA; Departments of Radiation Oncology (J.D.P.) and Neurosurgery (J.D.P.), The James Cancer Hospital at The Ohio State University Comprehensive Cancer Center, Columbus; Sunnybrook Health Sciences Centre (E.C.), University of Toronto, Canada; and Radiation Oncology (P.D.B.), Mayo Clinic, Rochester, MN
| | - Xiaoni Zhang
- From the Department of Neurology Bioland Laboratory (X.H., X. Zhang, X.R., Y.L., H.L., J.J., J.C., X. Zhuo, X.P., J.L., Z.Y., Y.T.) and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center (Y.T.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University; Guangdong Province Key Laboratory of Brain Function and Disease (Y.T.), Zhongshan School of Medicine, Sun Yat-Sen University; Department of Oncology (X.W.), The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Division of Radiation Oncology and Medical Sciences (M.L.K.C.), National Cancer Centre Singapore; Oncology Academic Programme (M.L.K.C.), Duke-NUS Medical School, Singapore; Department of Neurology (A.A.A.), Saint Andrew's State General Hospital of Patras, Greece; Neurological Clinic, Department of Experimental and Clinical Medicine (S.L.), Marche Polytechnic University, Italy; New York Proton Center (C.B.S.), New York; Thomas Jefferson University (J.G.), Philadelphia, PA; Departments of Radiation Oncology (J.D.P.) and Neurosurgery (J.D.P.), The James Cancer Hospital at The Ohio State University Comprehensive Cancer Center, Columbus; Sunnybrook Health Sciences Centre (E.C.), University of Toronto, Canada; and Radiation Oncology (P.D.B.), Mayo Clinic, Rochester, MN
| | - Xicheng Wang
- From the Department of Neurology Bioland Laboratory (X.H., X. Zhang, X.R., Y.L., H.L., J.J., J.C., X. Zhuo, X.P., J.L., Z.Y., Y.T.) and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center (Y.T.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University; Guangdong Province Key Laboratory of Brain Function and Disease (Y.T.), Zhongshan School of Medicine, Sun Yat-Sen University; Department of Oncology (X.W.), The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Division of Radiation Oncology and Medical Sciences (M.L.K.C.), National Cancer Centre Singapore; Oncology Academic Programme (M.L.K.C.), Duke-NUS Medical School, Singapore; Department of Neurology (A.A.A.), Saint Andrew's State General Hospital of Patras, Greece; Neurological Clinic, Department of Experimental and Clinical Medicine (S.L.), Marche Polytechnic University, Italy; New York Proton Center (C.B.S.), New York; Thomas Jefferson University (J.G.), Philadelphia, PA; Departments of Radiation Oncology (J.D.P.) and Neurosurgery (J.D.P.), The James Cancer Hospital at The Ohio State University Comprehensive Cancer Center, Columbus; Sunnybrook Health Sciences Centre (E.C.), University of Toronto, Canada; and Radiation Oncology (P.D.B.), Mayo Clinic, Rochester, MN
| | - Xiaoming Rong
- From the Department of Neurology Bioland Laboratory (X.H., X. Zhang, X.R., Y.L., H.L., J.J., J.C., X. Zhuo, X.P., J.L., Z.Y., Y.T.) and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center (Y.T.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University; Guangdong Province Key Laboratory of Brain Function and Disease (Y.T.), Zhongshan School of Medicine, Sun Yat-Sen University; Department of Oncology (X.W.), The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Division of Radiation Oncology and Medical Sciences (M.L.K.C.), National Cancer Centre Singapore; Oncology Academic Programme (M.L.K.C.), Duke-NUS Medical School, Singapore; Department of Neurology (A.A.A.), Saint Andrew's State General Hospital of Patras, Greece; Neurological Clinic, Department of Experimental and Clinical Medicine (S.L.), Marche Polytechnic University, Italy; New York Proton Center (C.B.S.), New York; Thomas Jefferson University (J.G.), Philadelphia, PA; Departments of Radiation Oncology (J.D.P.) and Neurosurgery (J.D.P.), The James Cancer Hospital at The Ohio State University Comprehensive Cancer Center, Columbus; Sunnybrook Health Sciences Centre (E.C.), University of Toronto, Canada; and Radiation Oncology (P.D.B.), Mayo Clinic, Rochester, MN
| | - Yi Li
- From the Department of Neurology Bioland Laboratory (X.H., X. Zhang, X.R., Y.L., H.L., J.J., J.C., X. Zhuo, X.P., J.L., Z.Y., Y.T.) and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center (Y.T.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University; Guangdong Province Key Laboratory of Brain Function and Disease (Y.T.), Zhongshan School of Medicine, Sun Yat-Sen University; Department of Oncology (X.W.), The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Division of Radiation Oncology and Medical Sciences (M.L.K.C.), National Cancer Centre Singapore; Oncology Academic Programme (M.L.K.C.), Duke-NUS Medical School, Singapore; Department of Neurology (A.A.A.), Saint Andrew's State General Hospital of Patras, Greece; Neurological Clinic, Department of Experimental and Clinical Medicine (S.L.), Marche Polytechnic University, Italy; New York Proton Center (C.B.S.), New York; Thomas Jefferson University (J.G.), Philadelphia, PA; Departments of Radiation Oncology (J.D.P.) and Neurosurgery (J.D.P.), The James Cancer Hospital at The Ohio State University Comprehensive Cancer Center, Columbus; Sunnybrook Health Sciences Centre (E.C.), University of Toronto, Canada; and Radiation Oncology (P.D.B.), Mayo Clinic, Rochester, MN
| | - Honghong Li
- From the Department of Neurology Bioland Laboratory (X.H., X. Zhang, X.R., Y.L., H.L., J.J., J.C., X. Zhuo, X.P., J.L., Z.Y., Y.T.) and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center (Y.T.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University; Guangdong Province Key Laboratory of Brain Function and Disease (Y.T.), Zhongshan School of Medicine, Sun Yat-Sen University; Department of Oncology (X.W.), The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Division of Radiation Oncology and Medical Sciences (M.L.K.C.), National Cancer Centre Singapore; Oncology Academic Programme (M.L.K.C.), Duke-NUS Medical School, Singapore; Department of Neurology (A.A.A.), Saint Andrew's State General Hospital of Patras, Greece; Neurological Clinic, Department of Experimental and Clinical Medicine (S.L.), Marche Polytechnic University, Italy; New York Proton Center (C.B.S.), New York; Thomas Jefferson University (J.G.), Philadelphia, PA; Departments of Radiation Oncology (J.D.P.) and Neurosurgery (J.D.P.), The James Cancer Hospital at The Ohio State University Comprehensive Cancer Center, Columbus; Sunnybrook Health Sciences Centre (E.C.), University of Toronto, Canada; and Radiation Oncology (P.D.B.), Mayo Clinic, Rochester, MN
| | - Jingru Jiang
- From the Department of Neurology Bioland Laboratory (X.H., X. Zhang, X.R., Y.L., H.L., J.J., J.C., X. Zhuo, X.P., J.L., Z.Y., Y.T.) and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center (Y.T.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University; Guangdong Province Key Laboratory of Brain Function and Disease (Y.T.), Zhongshan School of Medicine, Sun Yat-Sen University; Department of Oncology (X.W.), The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Division of Radiation Oncology and Medical Sciences (M.L.K.C.), National Cancer Centre Singapore; Oncology Academic Programme (M.L.K.C.), Duke-NUS Medical School, Singapore; Department of Neurology (A.A.A.), Saint Andrew's State General Hospital of Patras, Greece; Neurological Clinic, Department of Experimental and Clinical Medicine (S.L.), Marche Polytechnic University, Italy; New York Proton Center (C.B.S.), New York; Thomas Jefferson University (J.G.), Philadelphia, PA; Departments of Radiation Oncology (J.D.P.) and Neurosurgery (J.D.P.), The James Cancer Hospital at The Ohio State University Comprehensive Cancer Center, Columbus; Sunnybrook Health Sciences Centre (E.C.), University of Toronto, Canada; and Radiation Oncology (P.D.B.), Mayo Clinic, Rochester, MN
| | - Jinhua Cai
- From the Department of Neurology Bioland Laboratory (X.H., X. Zhang, X.R., Y.L., H.L., J.J., J.C., X. Zhuo, X.P., J.L., Z.Y., Y.T.) and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center (Y.T.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University; Guangdong Province Key Laboratory of Brain Function and Disease (Y.T.), Zhongshan School of Medicine, Sun Yat-Sen University; Department of Oncology (X.W.), The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Division of Radiation Oncology and Medical Sciences (M.L.K.C.), National Cancer Centre Singapore; Oncology Academic Programme (M.L.K.C.), Duke-NUS Medical School, Singapore; Department of Neurology (A.A.A.), Saint Andrew's State General Hospital of Patras, Greece; Neurological Clinic, Department of Experimental and Clinical Medicine (S.L.), Marche Polytechnic University, Italy; New York Proton Center (C.B.S.), New York; Thomas Jefferson University (J.G.), Philadelphia, PA; Departments of Radiation Oncology (J.D.P.) and Neurosurgery (J.D.P.), The James Cancer Hospital at The Ohio State University Comprehensive Cancer Center, Columbus; Sunnybrook Health Sciences Centre (E.C.), University of Toronto, Canada; and Radiation Oncology (P.D.B.), Mayo Clinic, Rochester, MN
| | - Xiaohuang Zhuo
- From the Department of Neurology Bioland Laboratory (X.H., X. Zhang, X.R., Y.L., H.L., J.J., J.C., X. Zhuo, X.P., J.L., Z.Y., Y.T.) and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center (Y.T.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University; Guangdong Province Key Laboratory of Brain Function and Disease (Y.T.), Zhongshan School of Medicine, Sun Yat-Sen University; Department of Oncology (X.W.), The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Division of Radiation Oncology and Medical Sciences (M.L.K.C.), National Cancer Centre Singapore; Oncology Academic Programme (M.L.K.C.), Duke-NUS Medical School, Singapore; Department of Neurology (A.A.A.), Saint Andrew's State General Hospital of Patras, Greece; Neurological Clinic, Department of Experimental and Clinical Medicine (S.L.), Marche Polytechnic University, Italy; New York Proton Center (C.B.S.), New York; Thomas Jefferson University (J.G.), Philadelphia, PA; Departments of Radiation Oncology (J.D.P.) and Neurosurgery (J.D.P.), The James Cancer Hospital at The Ohio State University Comprehensive Cancer Center, Columbus; Sunnybrook Health Sciences Centre (E.C.), University of Toronto, Canada; and Radiation Oncology (P.D.B.), Mayo Clinic, Rochester, MN
| | - Yaxuan Pi
- From the Department of Neurology Bioland Laboratory (X.H., X. Zhang, X.R., Y.L., H.L., J.J., J.C., X. Zhuo, X.P., J.L., Z.Y., Y.T.) and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center (Y.T.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University; Guangdong Province Key Laboratory of Brain Function and Disease (Y.T.), Zhongshan School of Medicine, Sun Yat-Sen University; Department of Oncology (X.W.), The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Division of Radiation Oncology and Medical Sciences (M.L.K.C.), National Cancer Centre Singapore; Oncology Academic Programme (M.L.K.C.), Duke-NUS Medical School, Singapore; Department of Neurology (A.A.A.), Saint Andrew's State General Hospital of Patras, Greece; Neurological Clinic, Department of Experimental and Clinical Medicine (S.L.), Marche Polytechnic University, Italy; New York Proton Center (C.B.S.), New York; Thomas Jefferson University (J.G.), Philadelphia, PA; Departments of Radiation Oncology (J.D.P.) and Neurosurgery (J.D.P.), The James Cancer Hospital at The Ohio State University Comprehensive Cancer Center, Columbus; Sunnybrook Health Sciences Centre (E.C.), University of Toronto, Canada; and Radiation Oncology (P.D.B.), Mayo Clinic, Rochester, MN
| | - Jinpeng Lin
- From the Department of Neurology Bioland Laboratory (X.H., X. Zhang, X.R., Y.L., H.L., J.J., J.C., X. Zhuo, X.P., J.L., Z.Y., Y.T.) and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center (Y.T.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University; Guangdong Province Key Laboratory of Brain Function and Disease (Y.T.), Zhongshan School of Medicine, Sun Yat-Sen University; Department of Oncology (X.W.), The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Division of Radiation Oncology and Medical Sciences (M.L.K.C.), National Cancer Centre Singapore; Oncology Academic Programme (M.L.K.C.), Duke-NUS Medical School, Singapore; Department of Neurology (A.A.A.), Saint Andrew's State General Hospital of Patras, Greece; Neurological Clinic, Department of Experimental and Clinical Medicine (S.L.), Marche Polytechnic University, Italy; New York Proton Center (C.B.S.), New York; Thomas Jefferson University (J.G.), Philadelphia, PA; Departments of Radiation Oncology (J.D.P.) and Neurosurgery (J.D.P.), The James Cancer Hospital at The Ohio State University Comprehensive Cancer Center, Columbus; Sunnybrook Health Sciences Centre (E.C.), University of Toronto, Canada; and Radiation Oncology (P.D.B.), Mayo Clinic, Rochester, MN
| | - Melvin L K Chua
- From the Department of Neurology Bioland Laboratory (X.H., X. Zhang, X.R., Y.L., H.L., J.J., J.C., X. Zhuo, X.P., J.L., Z.Y., Y.T.) and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center (Y.T.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University; Guangdong Province Key Laboratory of Brain Function and Disease (Y.T.), Zhongshan School of Medicine, Sun Yat-Sen University; Department of Oncology (X.W.), The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Division of Radiation Oncology and Medical Sciences (M.L.K.C.), National Cancer Centre Singapore; Oncology Academic Programme (M.L.K.C.), Duke-NUS Medical School, Singapore; Department of Neurology (A.A.A.), Saint Andrew's State General Hospital of Patras, Greece; Neurological Clinic, Department of Experimental and Clinical Medicine (S.L.), Marche Polytechnic University, Italy; New York Proton Center (C.B.S.), New York; Thomas Jefferson University (J.G.), Philadelphia, PA; Departments of Radiation Oncology (J.D.P.) and Neurosurgery (J.D.P.), The James Cancer Hospital at The Ohio State University Comprehensive Cancer Center, Columbus; Sunnybrook Health Sciences Centre (E.C.), University of Toronto, Canada; and Radiation Oncology (P.D.B.), Mayo Clinic, Rochester, MN
| | - Andreas A Argyriou
- From the Department of Neurology Bioland Laboratory (X.H., X. Zhang, X.R., Y.L., H.L., J.J., J.C., X. Zhuo, X.P., J.L., Z.Y., Y.T.) and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center (Y.T.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University; Guangdong Province Key Laboratory of Brain Function and Disease (Y.T.), Zhongshan School of Medicine, Sun Yat-Sen University; Department of Oncology (X.W.), The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Division of Radiation Oncology and Medical Sciences (M.L.K.C.), National Cancer Centre Singapore; Oncology Academic Programme (M.L.K.C.), Duke-NUS Medical School, Singapore; Department of Neurology (A.A.A.), Saint Andrew's State General Hospital of Patras, Greece; Neurological Clinic, Department of Experimental and Clinical Medicine (S.L.), Marche Polytechnic University, Italy; New York Proton Center (C.B.S.), New York; Thomas Jefferson University (J.G.), Philadelphia, PA; Departments of Radiation Oncology (J.D.P.) and Neurosurgery (J.D.P.), The James Cancer Hospital at The Ohio State University Comprehensive Cancer Center, Columbus; Sunnybrook Health Sciences Centre (E.C.), University of Toronto, Canada; and Radiation Oncology (P.D.B.), Mayo Clinic, Rochester, MN
| | - Simona Lattanzi
- From the Department of Neurology Bioland Laboratory (X.H., X. Zhang, X.R., Y.L., H.L., J.J., J.C., X. Zhuo, X.P., J.L., Z.Y., Y.T.) and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center (Y.T.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University; Guangdong Province Key Laboratory of Brain Function and Disease (Y.T.), Zhongshan School of Medicine, Sun Yat-Sen University; Department of Oncology (X.W.), The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Division of Radiation Oncology and Medical Sciences (M.L.K.C.), National Cancer Centre Singapore; Oncology Academic Programme (M.L.K.C.), Duke-NUS Medical School, Singapore; Department of Neurology (A.A.A.), Saint Andrew's State General Hospital of Patras, Greece; Neurological Clinic, Department of Experimental and Clinical Medicine (S.L.), Marche Polytechnic University, Italy; New York Proton Center (C.B.S.), New York; Thomas Jefferson University (J.G.), Philadelphia, PA; Departments of Radiation Oncology (J.D.P.) and Neurosurgery (J.D.P.), The James Cancer Hospital at The Ohio State University Comprehensive Cancer Center, Columbus; Sunnybrook Health Sciences Centre (E.C.), University of Toronto, Canada; and Radiation Oncology (P.D.B.), Mayo Clinic, Rochester, MN
| | - Charles B Simone
- From the Department of Neurology Bioland Laboratory (X.H., X. Zhang, X.R., Y.L., H.L., J.J., J.C., X. Zhuo, X.P., J.L., Z.Y., Y.T.) and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center (Y.T.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University; Guangdong Province Key Laboratory of Brain Function and Disease (Y.T.), Zhongshan School of Medicine, Sun Yat-Sen University; Department of Oncology (X.W.), The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Division of Radiation Oncology and Medical Sciences (M.L.K.C.), National Cancer Centre Singapore; Oncology Academic Programme (M.L.K.C.), Duke-NUS Medical School, Singapore; Department of Neurology (A.A.A.), Saint Andrew's State General Hospital of Patras, Greece; Neurological Clinic, Department of Experimental and Clinical Medicine (S.L.), Marche Polytechnic University, Italy; New York Proton Center (C.B.S.), New York; Thomas Jefferson University (J.G.), Philadelphia, PA; Departments of Radiation Oncology (J.D.P.) and Neurosurgery (J.D.P.), The James Cancer Hospital at The Ohio State University Comprehensive Cancer Center, Columbus; Sunnybrook Health Sciences Centre (E.C.), University of Toronto, Canada; and Radiation Oncology (P.D.B.), Mayo Clinic, Rochester, MN
| | - Jon Glass
- From the Department of Neurology Bioland Laboratory (X.H., X. Zhang, X.R., Y.L., H.L., J.J., J.C., X. Zhuo, X.P., J.L., Z.Y., Y.T.) and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center (Y.T.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University; Guangdong Province Key Laboratory of Brain Function and Disease (Y.T.), Zhongshan School of Medicine, Sun Yat-Sen University; Department of Oncology (X.W.), The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Division of Radiation Oncology and Medical Sciences (M.L.K.C.), National Cancer Centre Singapore; Oncology Academic Programme (M.L.K.C.), Duke-NUS Medical School, Singapore; Department of Neurology (A.A.A.), Saint Andrew's State General Hospital of Patras, Greece; Neurological Clinic, Department of Experimental and Clinical Medicine (S.L.), Marche Polytechnic University, Italy; New York Proton Center (C.B.S.), New York; Thomas Jefferson University (J.G.), Philadelphia, PA; Departments of Radiation Oncology (J.D.P.) and Neurosurgery (J.D.P.), The James Cancer Hospital at The Ohio State University Comprehensive Cancer Center, Columbus; Sunnybrook Health Sciences Centre (E.C.), University of Toronto, Canada; and Radiation Oncology (P.D.B.), Mayo Clinic, Rochester, MN
| | - Joshua D Palmer
- From the Department of Neurology Bioland Laboratory (X.H., X. Zhang, X.R., Y.L., H.L., J.J., J.C., X. Zhuo, X.P., J.L., Z.Y., Y.T.) and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center (Y.T.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University; Guangdong Province Key Laboratory of Brain Function and Disease (Y.T.), Zhongshan School of Medicine, Sun Yat-Sen University; Department of Oncology (X.W.), The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Division of Radiation Oncology and Medical Sciences (M.L.K.C.), National Cancer Centre Singapore; Oncology Academic Programme (M.L.K.C.), Duke-NUS Medical School, Singapore; Department of Neurology (A.A.A.), Saint Andrew's State General Hospital of Patras, Greece; Neurological Clinic, Department of Experimental and Clinical Medicine (S.L.), Marche Polytechnic University, Italy; New York Proton Center (C.B.S.), New York; Thomas Jefferson University (J.G.), Philadelphia, PA; Departments of Radiation Oncology (J.D.P.) and Neurosurgery (J.D.P.), The James Cancer Hospital at The Ohio State University Comprehensive Cancer Center, Columbus; Sunnybrook Health Sciences Centre (E.C.), University of Toronto, Canada; and Radiation Oncology (P.D.B.), Mayo Clinic, Rochester, MN
| | - Edward Chow
- From the Department of Neurology Bioland Laboratory (X.H., X. Zhang, X.R., Y.L., H.L., J.J., J.C., X. Zhuo, X.P., J.L., Z.Y., Y.T.) and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center (Y.T.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University; Guangdong Province Key Laboratory of Brain Function and Disease (Y.T.), Zhongshan School of Medicine, Sun Yat-Sen University; Department of Oncology (X.W.), The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Division of Radiation Oncology and Medical Sciences (M.L.K.C.), National Cancer Centre Singapore; Oncology Academic Programme (M.L.K.C.), Duke-NUS Medical School, Singapore; Department of Neurology (A.A.A.), Saint Andrew's State General Hospital of Patras, Greece; Neurological Clinic, Department of Experimental and Clinical Medicine (S.L.), Marche Polytechnic University, Italy; New York Proton Center (C.B.S.), New York; Thomas Jefferson University (J.G.), Philadelphia, PA; Departments of Radiation Oncology (J.D.P.) and Neurosurgery (J.D.P.), The James Cancer Hospital at The Ohio State University Comprehensive Cancer Center, Columbus; Sunnybrook Health Sciences Centre (E.C.), University of Toronto, Canada; and Radiation Oncology (P.D.B.), Mayo Clinic, Rochester, MN
| | - Paul D Brown
- From the Department of Neurology Bioland Laboratory (X.H., X. Zhang, X.R., Y.L., H.L., J.J., J.C., X. Zhuo, X.P., J.L., Z.Y., Y.T.) and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center (Y.T.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University; Guangdong Province Key Laboratory of Brain Function and Disease (Y.T.), Zhongshan School of Medicine, Sun Yat-Sen University; Department of Oncology (X.W.), The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Division of Radiation Oncology and Medical Sciences (M.L.K.C.), National Cancer Centre Singapore; Oncology Academic Programme (M.L.K.C.), Duke-NUS Medical School, Singapore; Department of Neurology (A.A.A.), Saint Andrew's State General Hospital of Patras, Greece; Neurological Clinic, Department of Experimental and Clinical Medicine (S.L.), Marche Polytechnic University, Italy; New York Proton Center (C.B.S.), New York; Thomas Jefferson University (J.G.), Philadelphia, PA; Departments of Radiation Oncology (J.D.P.) and Neurosurgery (J.D.P.), The James Cancer Hospital at The Ohio State University Comprehensive Cancer Center, Columbus; Sunnybrook Health Sciences Centre (E.C.), University of Toronto, Canada; and Radiation Oncology (P.D.B.), Mayo Clinic, Rochester, MN
| | - Zongwei Yue
- From the Department of Neurology Bioland Laboratory (X.H., X. Zhang, X.R., Y.L., H.L., J.J., J.C., X. Zhuo, X.P., J.L., Z.Y., Y.T.) and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center (Y.T.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University; Guangdong Province Key Laboratory of Brain Function and Disease (Y.T.), Zhongshan School of Medicine, Sun Yat-Sen University; Department of Oncology (X.W.), The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Division of Radiation Oncology and Medical Sciences (M.L.K.C.), National Cancer Centre Singapore; Oncology Academic Programme (M.L.K.C.), Duke-NUS Medical School, Singapore; Department of Neurology (A.A.A.), Saint Andrew's State General Hospital of Patras, Greece; Neurological Clinic, Department of Experimental and Clinical Medicine (S.L.), Marche Polytechnic University, Italy; New York Proton Center (C.B.S.), New York; Thomas Jefferson University (J.G.), Philadelphia, PA; Departments of Radiation Oncology (J.D.P.) and Neurosurgery (J.D.P.), The James Cancer Hospital at The Ohio State University Comprehensive Cancer Center, Columbus; Sunnybrook Health Sciences Centre (E.C.), University of Toronto, Canada; and Radiation Oncology (P.D.B.), Mayo Clinic, Rochester, MN
| | - Yamei Tang
- From the Department of Neurology Bioland Laboratory (X.H., X. Zhang, X.R., Y.L., H.L., J.J., J.C., X. Zhuo, X.P., J.L., Z.Y., Y.T.) and Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center (Y.T.), Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University; Guangdong Province Key Laboratory of Brain Function and Disease (Y.T.), Zhongshan School of Medicine, Sun Yat-Sen University; Department of Oncology (X.W.), The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China; Division of Radiation Oncology and Medical Sciences (M.L.K.C.), National Cancer Centre Singapore; Oncology Academic Programme (M.L.K.C.), Duke-NUS Medical School, Singapore; Department of Neurology (A.A.A.), Saint Andrew's State General Hospital of Patras, Greece; Neurological Clinic, Department of Experimental and Clinical Medicine (S.L.), Marche Polytechnic University, Italy; New York Proton Center (C.B.S.), New York; Thomas Jefferson University (J.G.), Philadelphia, PA; Departments of Radiation Oncology (J.D.P.) and Neurosurgery (J.D.P.), The James Cancer Hospital at The Ohio State University Comprehensive Cancer Center, Columbus; Sunnybrook Health Sciences Centre (E.C.), University of Toronto, Canada; and Radiation Oncology (P.D.B.), Mayo Clinic, Rochester, MN.
| |
Collapse
|
207
|
An Iatrogenic Model of Brain Small-Vessel Disease: Post-Radiation Encephalopathy. Int J Mol Sci 2020; 21:ijms21186506. [PMID: 32899565 PMCID: PMC7555594 DOI: 10.3390/ijms21186506] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/27/2020] [Accepted: 09/01/2020] [Indexed: 12/21/2022] Open
Abstract
We studied 114 primitive cerebral neoplasia, that were surgically treated, and underwent radiotherapy (RT), and compared their results to those obtained by 190 patients diagnosed with subcortical vascular dementia (sVAD). Patients with any form of primitive cerebral neoplasia underwent whole-brain radiotherapy. All the tumor patients had regional field partial brain RT, which encompassed each tumor, with an average margin of 2.6 cm from the initial target tumor volume. We observed in our patients who have been exposed to a higher dose of RT (30–65 Gy) a cognitive and behavior decline similar to that observed in sVAD, with the frontal dysexecutive syndrome, apathy, and gait alterations, but with a more rapid onset and with an overwhelming effect. Multiple mechanisms are likely to be involved in radiation-induced cognitive impairment. The active site of RT brain damage is the white matter areas, particularly the internal capsule, basal ganglia, caudate, hippocampus, and subventricular zone. In all cases, radiation damage inside the brain mainly focuses on the cortical–subcortical frontal loops, which integrate and process the flow of information from the cortical areas, where executive functions are “elaborated” and prepared, towards the thalamus, subthalamus, and cerebellum, where they are continuously refined and executed. The active mechanisms that RT drives are similar to those observed in cerebral small vessel disease (SVD), leading to sVAD. The RT’s primary targets, outside the tumor mass, are the blood–brain barrier (BBB), the small vessels, and putative mechanisms that can be taken into account are oxidative stress and neuro-inflammation, strongly associated with the alteration of NMDA receptor subunit composition.
Collapse
|
208
|
Helis CA, Hughes RT, Glenn CW, Lanier CM, Masters AH, Dohm A, Ahmed T, Ruiz J, Triozzi P, Gondal H, Cramer CK, Tatter SB, Laxton AW, Xing F, Lo HW, Su J, Watabe K, Wang G, Whitlow CT, Chan MD. Predictors of Adverse Radiation Effect in Brain Metastasis Patients Treated With Stereotactic Radiosurgery and Immune Checkpoint Inhibitor Therapy. Int J Radiat Oncol Biol Phys 2020; 108:295-303. [DOI: 10.1016/j.ijrobp.2020.06.057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 12/19/2022]
|
209
|
Constanzo J, Midavaine É, Fouquet J, Lepage M, Descoteaux M, Kirby K, Tremblay L, Masson-Côté L, Geha S, Longpré JM, Paquette B, Sarret P. Brain irradiation leads to persistent neuroinflammation and long-term neurocognitive dysfunction in a region-specific manner. Prog Neuropsychopharmacol Biol Psychiatry 2020; 102:109954. [PMID: 32360786 DOI: 10.1016/j.pnpbp.2020.109954] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/14/2020] [Accepted: 04/28/2020] [Indexed: 01/04/2023]
Abstract
Long-term cognitive deficits are observed after treatment of brain tumors or metastases by radiotherapy. Treatment optimization thus requires a better understanding of the effects of radiotherapy on specific brain regions, according to their sensitivity and interconnectivity. In the present study, behavioral tests supported by immunohistology and magnetic resonance imaging provided a consistent picture of the persistent neurocognitive decline and neuroinflammation after the onset of irradiation-induced necrosis in the right primary somatosensory cortex of Fischer rats. Necrosis surrounded by neovascularization was first detected 54 days after irradiation and then spread to 110 days in the primary motor cortex, primary somatosensory region, striatum and right ventricle, resulting in fiber bundle disruption and demyelination in the corpus callosum of the right hemisphere. These structural damages translated into selective behavioral changes including spatial memory loss, disinhibition of anxiety-like behaviors, hyperactivity and pain hypersensitivity, but no significant alteration in motor coordination and grip strength abilities. Concomitantly, activated microglia and reactive astrocytes, accompanied by infiltration of leukocytes (CD45+) and T-cells (CD3+) cooperated to shape the neuroinflammation response. Overall, our study suggests that the slow and gradual onset of cellular damage would allow adaptation in brain regions that are susceptible to neuronal plasticity; while other cerebral structures that do not have this capacity would be more affected. The planning of radiotherapy, adjusted to the sensitivity and adaptability of brain structures, could therefore preserve certain neurocognitive functions; while higher doses of radiation could be delivered to brain areas that can better adapt to this treatment. In addition, strategies to block early post-radiation events need to be explored to prevent the development of long-term cognitive dysfunction.
Collapse
Affiliation(s)
- Julie Constanzo
- Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Élora Midavaine
- Department of Pharmacology-Physiology, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Jérémie Fouquet
- Sherbrooke Molecular Imaging Center, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Martin Lepage
- Sherbrooke Molecular Imaging Center, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Maxime Descoteaux
- Computer Science Department, Université de Sherbrooke, Sherbrooke, Québec J1K 2R1, Canada
| | - Karyn Kirby
- Department of Pharmacology-Physiology, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Luc Tremblay
- Sherbrooke Molecular Imaging Center, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Laurence Masson-Côté
- Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada; Service of Radiation Oncology, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Sameh Geha
- Department of Pathology, Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Jean-Michel Longpré
- Department of Pharmacology-Physiology, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Benoit Paquette
- Center for Research in Radiotherapy, Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada.
| | - Philippe Sarret
- Department of Pharmacology-Physiology, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada.
| |
Collapse
|
210
|
Wu A, Jin MC, Meola A, Wong HN, Chang SD. Efficacy and toxicity of particle radiotherapy in WHO grade II and grade III meningiomas: a systematic review. Neurosurg Focus 2020; 46:E12. [PMID: 31153145 DOI: 10.3171/2019.3.focus1967] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 03/25/2019] [Indexed: 11/06/2022]
Abstract
OBJECTIVEAdjuvant radiotherapy has become a common addition to the management of high-grade meningiomas, as immediate treatment with radiation following resection has been associated with significantly improved outcomes. Recent investigations into particle therapy have expanded into the management of high-risk meningiomas. Here, the authors systematically review studies on the efficacy and utility of particle-based radiotherapy in the management of high-grade meningioma.METHODSA literature search was developed by first defining the population, intervention, comparison, outcomes, and study design (PICOS). A search strategy was designed for each of three electronic databases: PubMed, Embase, and Scopus. Data extraction was conducted in accordance with the PRISMA guidelines. Outcomes of interest included local disease control, overall survival, and toxicity, which were compared with historical data on photon-based therapies.RESULTSEleven retrospective studies including 240 patients with atypical (WHO grade II) and anaplastic (WHO grade III) meningioma undergoing particle radiation therapy were identified. Five of the 11 studies included in this systematic review focused specifically on WHO grade II and III meningiomas; the others also included WHO grade I meningioma. Across all of the studies, the median follow-up ranged from 6 to 145 months. Local control rates for high-grade meningiomas ranged from 46.7% to 86% by the last follow-up or at 5 years. Overall survival rates ranged from 0% to 100% with better prognoses for atypical than for malignant meningiomas. Radiation necrosis was the most common adverse effect of treatment, occurring in 3.9% of specified cases.CONCLUSIONSDespite the lack of randomized prospective trials, this review of existing retrospective studies suggests that particle therapy, whether an adjuvant or a stand-alone treatment, confers survival benefit with a relatively low risk for severe treatment-derived toxicity compared to standard photon-based therapy. However, additional controlled studies are needed.
Collapse
Affiliation(s)
- Adela Wu
- 1Department of Neurosurgery, Stanford Health Care, Palo Alto
| | - Michael C Jin
- 2Stanford University School of Medicine, Stanford; and
| | - Antonio Meola
- 1Department of Neurosurgery, Stanford Health Care, Palo Alto
| | - Hong-Nei Wong
- 3Lane Medical Library, Stanford Medicine, Palo Alto, California
| | - Steven D Chang
- 1Department of Neurosurgery, Stanford Health Care, Palo Alto
| |
Collapse
|
211
|
Mitchell TJ, Seitzman BA, Ballard N, Petersen SE, Shimony JS, Leuthardt EC. Human Brain Functional Network Organization Is Disrupted After Whole-Brain Radiation Therapy. Brain Connect 2020; 10:29-38. [PMID: 31964163 DOI: 10.1089/brain.2019.0713] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Radiation therapy (RT) plays a vital role in the treatment of brain cancers, but it frequently results in cognitive decline in the patients who receive it. Because the underlying mechanisms for this decline remain poorly understood, the brain is typically treated as a single, uniform volume when evaluating the toxic effects of RT plans. This ignorance represents a significant deficit in the field of radiation oncology, as the technology exists to manipulate dose distributions to spare regions of the brain, but there exists no body of knowledge regarding what is critical to spare. This deficit exists due to the numerous confounding factors that are frequently associated with radiotherapy, including the tumors themselves, other treatments such as surgery and chemotherapy, and dose gradients across the brain. Here, we present a case in which a 57-year-old male patient received a uniform dose of radiation across the whole brain, did not receive concurrent chemotherapy, had minimal surgical intervention and a small tumor burden, and received resting-state functional magnetic resonance imaging (fMRI) scans both before and after RT. To our knowledge, this is the first study on the effects of whole-brain radiotherapy on functional network organization, and this patient's treatment regimen represents a rare and non-replicable opportunity to isolate the effects of radiation on functional connectivity. We observed substantial changes in the subject's behavior and functional network organization over a 12-month timeframe. Interestingly, the homogenous radiation dose to the brain had a heterogeneous effect on cortical networks, and the functional networks most affected correspond with observed cognitive behavioral deficits. This novel study suggests that the cognitive decline that occurs after whole-brain radiation therapy may be network specific and related to the disruption of large-scale distributed functional systems, and it indicates that fMRI is a promising avenue of study for optimizing cognitive outcomes after RT.
Collapse
Affiliation(s)
- Timothy J Mitchell
- Department of Radiation Oncology and Washington University in St. Louis-School of Medicine, St. Louis, Missouri
| | - Benjamin A Seitzman
- Department of Neurology, Washington University in St. Louis-School of Medicine, St. Louis, Missouri
| | - Nicholas Ballard
- Department of Radiation Oncology and Washington University in St. Louis-School of Medicine, St. Louis, Missouri
| | - Steven E Petersen
- Department of Neurology, Washington University in St. Louis-School of Medicine, St. Louis, Missouri.,Mallinckrodt Institute of Radiology, Washington University in St. Louis-School of Medicine, St. Louis, Missouri.,Department of Neuroscience, Washington University in St. Louis-School of Medicine, St. Louis, Missouri.,Department of Biomedical Engineering, Washington University in St. Louis-School of Engineering and Applied Science, St. Louis, Missouri.,Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, Missouri
| | - Joshua S Shimony
- Mallinckrodt Institute of Radiology, Washington University in St. Louis-School of Medicine, St. Louis, Missouri
| | - Eric C Leuthardt
- Department of Neuroscience, Washington University in St. Louis-School of Medicine, St. Louis, Missouri.,Department of Biomedical Engineering, Washington University in St. Louis-School of Engineering and Applied Science, St. Louis, Missouri.,Department of Mechanical Engineering and Materials Science, Washington University in St. Louis-School of Engineering and Applied Science, St. Louis, Missouri.,Center for Innovation in Neuroscience and Technology and Washington University in St. Louis-School of Medicine, St. Louis, Missouri.,Brain Laser Center, Washington University in St. Louis-School of Medicine, St. Louis, Missouri.,Department of Neurological Surgery, Washington University in St. Louis-School of Medicine, St. Louis, Missouri
| |
Collapse
|
212
|
Chargari C, Supiot S, Hennequin C, Chapel A, Simon JM. [Treatment of radiation-induced late effects: What's new?]. Cancer Radiother 2020; 24:602-611. [PMID: 32855027 DOI: 10.1016/j.canrad.2020.06.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 05/29/2020] [Accepted: 06/07/2020] [Indexed: 02/06/2023]
Abstract
Mechanisms of late radio-induced lesions are the result of multiple and complex phenomena, with many entangled cellular and tissue factors. The biological continuum between acute and late radio-induced effects will be described, with firstly a break in homeostasis that leads to cellular redistributions. New insights into late toxicity will finally be addressed. Individual radiosensitivity is a primary factor for the development of late toxicity, and clinicians urgently need predictive tests to offer truly personalized radiation therapy. An update will be made on the various functional and genetic tests currently being validated. The management of radio-induced side effects remains a frequent issue for radiation oncologists, and an update will be made for certain specific clinical situations. Finally, an innovative management for patients with significant side effects after pelvic radiotherapy will be developed, involved mesenchymal stem cell transplantation, with the presentation of the "PRISME" protocol currently open to patients recruitment.
Collapse
Affiliation(s)
- C Chargari
- Département de radiothérapie, Gustave-Roussy Cancer Campus, 114, rue Édouard-Vaillant, 94800 Villejuif France
| | - S Supiot
- Département d'oncologie radiothérapie, institut de cancérologie de l'ouest - centre René-Gauducheau, boulevard Jacques-Monod, 44805 Saint-Herblain cedex, France; Institut de recherche en santé de l'université de Nantes, université de Nantes, 8, quai Moncousu, BP 70721, 44007 Nantes cedex 1, France; Inserm, U1232 Centre de recherche en cancérologie et immunologie de Nantes - Angers (CRCINA), 8, quai Moncousu, BP 70721, 44007 Nantes cedex 1, France; CNRS, ERL 6001, 8, quai Moncousu, BP 70721, 44007 Nantes cedex 1, France
| | - C Hennequin
- Service de cancérologie-radiothérapie, hôpital Saint-Louis, 1, avenue Claude-Vellefeaux, 75475 Paris, France
| | - A Chapel
- Service de recherche en radiobiologie et en médecine régénérative, laboratoire de radiobiologie des expositions médicales, Institut de radioprotection et de sûreté nucléaire (IRSN), 31, avenue de la Division-Leclerc, 92260 Fontenay-aux-Roses, France
| | - J-M Simon
- Sorbonne université, 21, rue de l'École-de-Médecine, 75006 Paris, France; Service d'oncologie radiothérapie, hôpital Pitié-Salpêtrière, AP-HP, 47-83, boulevard de l'Hôpital, 75651 Paris cedex 13, France.
| |
Collapse
|
213
|
Raschke F, Seidlitz A, Wesemann T, Löck S, Jentsch C, Platzek I, Petr J, van den Hoff J, Kotzerke J, Beuthien-Baumann B, Baumann M, Linn J, Krause M, Troost EGC. Dose dependent cerebellar atrophy in glioma patients after radio(chemo)therapy. Radiother Oncol 2020; 150:262-267. [PMID: 32739316 DOI: 10.1016/j.radonc.2020.07.044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/21/2020] [Accepted: 07/27/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND PURPOSE Radiotherapy is a standard treatment option for high-grade gliomas. Brain atrophy has previously been associated with radiotherapy. The goal of this study was to investigate dose dependent cerebellar atrophy using prospective, longitudinal MR data from adult glioma patients who received radiotherapy. MATERIALS AND METHODS Cerebellar volumes were measured using T1-weighted MR images from 91 glioma patients before radiotherapy (N = 91) and from longitudinal follow-ups acquired in three monthly intervals (N = 349). Relative cerebellar volumes were calculated as ratios to the corresponding baseline values. Univariate mixed effects models were used to determine factors that were significantly associated with relative cerebellar volumes. These factors were subsequently included as fixed effects in a final multivariate linear mixed effects model. RESULTS In multivariate analysis, cerebellar volume decreased significantly as a function of time (p < 0.001), time × dose (p < 0.001) and patient age (p = 0.007). Considering a 55 year patient receiving a mean cerebellar dose of 0 Gy (10 Gy), the linear mixed effects model predicts a relative cerebellar volume loss of 0.4% (2.0%) after 1 year and 0.7% (3.6%) after 2 years. Compared to patients treated with photons, the cerebellar dose was significantly lower in patients treated with proton therapy (p < 0.001, r = 0.62). CONCLUSION Cerebellar volume decreased significantly and irreversibly after radiotherapy as function of time and mean cerebellar dose. Further work is now needed to correlate these results with cognitive function and motor performance.
Collapse
Affiliation(s)
- Felix Raschke
- Institute of Radiooncology - OncoRay, Helmholtz-Zentrum Dresden-Rossendorf, Germany; OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany.
| | - Annekatrin Seidlitz
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Tim Wesemann
- Institute of Neuroradiology, University Hospital Carl Gustav Carus and Medical Faculty of Technische Universität, Dresden, Germany
| | - Steffen Löck
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christina Jentsch
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Ivan Platzek
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Department of Diagnostic and Interventional Radiology, Germany
| | - Jan Petr
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiopharmaceutical Cancer Research, Center for Positron Emission Tomography, Germany
| | - Jörg van den Hoff
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiopharmaceutical Cancer Research, Center for Positron Emission Tomography, Germany
| | - Jörg Kotzerke
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Department of Nuclear Medicine, Germany
| | - Bettina Beuthien-Baumann
- Heidelberg Institute for Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), Germany; Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Baumann
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), Partner Site, Heidelberg, Germany
| | - Jennifer Linn
- Institute of Neuroradiology, University Hospital Carl Gustav Carus and Medical Faculty of Technische Universität, Dresden, Germany
| | - Mechthild Krause
- Institute of Radiooncology - OncoRay, Helmholtz-Zentrum Dresden-Rossendorf, Germany; OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and; Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Esther G C Troost
- Institute of Radiooncology - OncoRay, Helmholtz-Zentrum Dresden-Rossendorf, Germany; OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and; Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| |
Collapse
|
214
|
Neumann S, Porritt MJ, Osman AM, Kuhn HG. Cranial irradiation at early postnatal age impairs stroke-induced neural stem/progenitor cell response in the adult brain. Sci Rep 2020; 10:12369. [PMID: 32703986 PMCID: PMC7378832 DOI: 10.1038/s41598-020-69266-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 07/09/2020] [Indexed: 11/29/2022] Open
Abstract
Cranial irradiation (IR) is commonly used to treat primary brain tumors and metastatic diseases. However, cranial IR-treated patients often develop vascular abnormalities later in life that increase their risk for cerebral ischemia. Studies in rodents have demonstrated that IR impairs maintenance of the neural stem/precursor cell (NSPC) pool and depletes neurogenesis. We and others have previously shown that stroke triggers NSPC proliferation in the subventricular zone and migration towards the stroke-injured neocortex. Whether this response is sustained in the irradiated brain remains unknown. Here, we demonstrate that cranial IR in mice at an early postnatal age significantly reduced the number to neuronal progenitors responding to cortical stroke in adults. This was accompanied by a reduced number of microglia/macrophages in the peri-infarct cortex; however, the astrocytic response was not altered. Our findings indicate that IR impairs the endogenous repair capacity in the brain in response to stroke, hence pointing to another side effect of cranial radiotherapy which requires further attention.
Collapse
Affiliation(s)
- Susanne Neumann
- Department of Clinical Neuroscience, Institute for Neuroscience and Physiology, University of Gothenburg, Box 436, 405 30, Gothenburg, Sweden.,Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, 171 76, Stockholm, Sweden
| | - Michelle J Porritt
- Department of Clinical Neuroscience, Institute for Neuroscience and Physiology, University of Gothenburg, Box 436, 405 30, Gothenburg, Sweden
| | - Ahmed M Osman
- Department of Women's and Children's Health, Karolinska Institutet, 171 64, Stockholm, Sweden
| | - H Georg Kuhn
- Department of Clinical Neuroscience, Institute for Neuroscience and Physiology, University of Gothenburg, Box 436, 405 30, Gothenburg, Sweden.
| |
Collapse
|
215
|
Eley JG, Chadha AS, Quini C, Vichaya EG, Zhang C, Davis J, Sahoo N, Waddell J, Leiser D, Dilmanian FA, Krishnan S. Pilot study of neurologic toxicity in mice after proton minibeam therapy. Sci Rep 2020; 10:11368. [PMID: 32647361 PMCID: PMC7347840 DOI: 10.1038/s41598-020-68015-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 04/24/2020] [Indexed: 01/03/2023] Open
Abstract
Proton minibeams (MBs) comprised of parallel planar beamlets were evaluated for their ability to spare healthy brain compared to proton broad beams (BBs). Juvenile mice were given partial brain irradiation of 10 or 30 Gy integral dose using 100 MeV protons configured either as BBs or arrays of 0.3-mm planar MBs spaced 1.0 mm apart on center. Neurologic toxicity was evaluated during an 8-month surveillance: no overt constitutional or neurologic dysfunction was noted for any study animals. Less acute epilation was observed in MB than BB mice. Persistent chronic inflammation was noted along the entire BB path in BB mice whereas inflammation was confined to just within the MB peak regions in MB mice. The potential neurologic sparing, possibly via reduced volume of chronic inflammation, offers a compelling rationale for clinical advancement of this proton technique.
Collapse
Affiliation(s)
- John G Eley
- Department of Radiation Oncology, Vanderbilt University School of Medicine, 2220 Pierce Avenue, Preston Research Building B1003, Nashville, TN, 37232, USA.
| | - Awalpreet S Chadha
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Caio Quini
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Elisabeth G Vichaya
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Cancan Zhang
- Department of Radiation Oncology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA
| | - James Davis
- Department of Pathology, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY, 11794, USA
| | - Narayan Sahoo
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Jaylyn Waddell
- Department of Pediatrics, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA
| | - Dominic Leiser
- Department of Radiation Oncology, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA
| | - F Avraham Dilmanian
- Departments of Radiology, Neurology, and Radiation Oncology, Stony Brook Medicine, 101 Nicolls Road, Stony Brook, NY, 11794, USA
| | - Sunil Krishnan
- Department of Radiation Oncology, Mayo Clinic Florida, 4500 San Pablo Road S., Jacksonville, FL, 32224, USA.
| |
Collapse
|
216
|
Microglia Purinoceptor P2Y6: An Emerging Therapeutic Target in CNS Diseases. Cells 2020; 9:cells9071595. [PMID: 32630251 PMCID: PMC7407337 DOI: 10.3390/cells9071595] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/24/2020] [Accepted: 06/29/2020] [Indexed: 12/14/2022] Open
Abstract
The purinergic receptor P2Y6 is expressed in immune cells, including the microglia that are implicated in neurological disorders. Its ligand, UDP, is a signaling molecule that can serve as an “find-me” signal when released in significant quantities by damaged/dying cells. The binding of UDP by P2Y6R leads to the activation of different biochemical pathways, depending on the disease context and the pathological environment. Generally, P2Y6R stimulates phagocytosis. However, whether or not phagocytosis coincides with cell activation or the secretion of pro-inflammatory cytokines needs further investigation. The current review aims to discuss the various functions of P2Y6R in some CNS disorders. We present evidence that P2Y6R may have a detrimental or beneficial role in the nervous system, in the context of neurological pathologies, such as ischemic stroke, Alzheimer’s disease, Parkinson’s disease, radiation-induced brain injury, and neuropathic pain.
Collapse
|
217
|
Sultana N, Sun C, Katsube T, Wang B. Biomarkers of Brain Damage Induced by Radiotherapy. Dose Response 2020; 18:1559325820938279. [PMID: 32694960 PMCID: PMC7350401 DOI: 10.1177/1559325820938279] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/26/2020] [Accepted: 06/05/2020] [Indexed: 12/18/2022] Open
Abstract
Radiotherapy remains currently a critical component for both primary and metastatic brain tumors either alone or in combination with surgery, chemotherapy, and molecularly targeted agents, while it could cause simultaneously normal brain tissue injury leading to serious health consequences, that is, development of cognitive impairments following cranial radiotherapy is considered as a critical clinical disadvantage especially for the whole brain radiotherapy. Biomarkers can help to detect the accurate physiology or conditions of patients with brain tumor and develop effective treatment procedures for these patients. In the near future, biomarkers will become one of the prime driving forces of cancer treatment. In this minireview, we analyze the documented work on the acute brain damage and late consequences induced by radiotherapy, identify the biomarkers, in particular, the predictive biomarkers for the damage, and summarize the biological significance of the biomarkers. It is expected that translation of these research advance to radiotherapy would assist stratifying patients for optimized treatment and improving therapeutic efficacy and the quality of life.
Collapse
Affiliation(s)
- Nahida Sultana
- Institute of Food and Radiation Biology, Atomic Energy Research Establishment, Bangladesh Atomic Energy Commission, Dhaka, People’s Republic of Bangladesh
| | - Chao Sun
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, People’s Republic of China
| | - Takanori Katsube
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Bing Wang
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| |
Collapse
|
218
|
Parente A, Maciel ES, J O Dierckx RA, Langendijk JA, de Vries EFJ, Doorduin J. Delayed effects of a single-dose whole-brain radiation therapy on glucose metabolism and myelin density: a longitudinal PET study. Int J Radiat Biol 2020; 96:1135-1143. [PMID: 32602390 DOI: 10.1080/09553002.2020.1787542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
PURPOSE Radiotherapy is an important treatment option for brain tumors, but the unavoidable irradiation of normal brain tissue can lead to delayed cognitive impairment. The mechanisms involved are still not well explained and, therefore, new tools to investigate the processes leading to the delayed symptoms of brain irradiation are warranted. In this study, positron emission tomography (PET) is used to explore delayed functional changes induced by brain irradiation. MATERIALS AND METHODS Male Wistar rats were subjected to a single 25-Gy dose of whole brain X-ray irradiation, or sham-irradiation. To investigate delayed effects of radiation on cerebral glucose metabolism and myelin density, 18F-fluorodeoxyglucose (18F-FDG) PET scans were performed at baseline and on day 64 and 94, whereas N-11C-methyl-4,4'-diaminostilbene (11C-MeDAS) PET scans were performed at baseline and on day 60 and 90 post-irradiation. In addition, the open field test (OFT) and novel spatial recognition (NSR) test were performed at baseline and on days 59 and 89 to investigate whether whole brain irradiation induces behavioral changes. RESULTS Whole-brain irradiation caused loss of bodyweight and delayed cerebral hypometabolism, with 18F-FDG uptake in all brain regions being significantly decreased in irradiated rat on day 64 while it remained unchanged in control animals. Only amygdala and cortical brain regions of irradiated rats still showed reduced 18F-FDG uptake on day 94. 11C-MeDAS uptake in control animals was significantly lower on days 60 and 90 than at the baseline, suggesting a reduction in myelin density in young adults. In irradiated animals, 11C-MeDAS uptake was similarly reduced on day 60, but on day 90 tracer uptake was somewhat increased and not significantly different from baseline anymore. Behavioral tests showed a similar pattern in control and irradiated animals. In both groups, the OFT showed significantly reduced mobility on days 59 and 89, whereas the NSR did not reveal any significant changes in spatial memory over time. Interestingly, a positive correlation between the NSR and 11C-MeDAS uptake was observed in irradiated rats. CONCLUSIONS Whole-brain irradiation causes delayed brain hypometabolism, which is not accompanied by white matter loss. Irradiated animals showed similar behavioral changes over time as control animals and, therefore, cerebral hypometabolism could not be linked to behavioral abnormalities. However, spatial memory seems to be associated with myelin density in irradiated rats.
Collapse
Affiliation(s)
- Andrea Parente
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Elisa Scandiuzzi Maciel
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Johannes A Langendijk
- Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Erik F J de Vries
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
219
|
Cayuela N, Jaramillo-Jiménez E, Càmara E, Majós C, Vidal N, Lucas A, Gil-Gil M, Graus F, Bruna J, Simó M. Cognitive and brain structural changes in long-term oligodendroglial tumor survivors. Neuro Oncol 2020; 21:1470-1479. [PMID: 31549152 DOI: 10.1093/neuonc/noz130] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND We identify cognitive impairment and MRI structural brain changes in long-term oligodendroglial tumor survivors treated with radiation therapy (RT) alone (21%) or with chemotherapy (CT) (79%). METHODS Oligodendroglial tumor patients (based on the World Health Organization [WHO] 2007 classification) who completed RT ± CT at least 2 years before the study initiation, were classified into 3 groups according to the time treatment was completed: Group 1 = 2-5 years (n = 22), Group 2 = 6-10 years (n = 13), and Group 3 >10 years (n = 13). All patients had a cross-sectional neuropsychological evaluation (n = 48) and a longitudinal volumetric analysis (gray matter [GM; n = 34]) between postsurgical and last follow-up MRI. White matter (WM) changes on MRI were assessed using a qualitative scale. RESULTS There were no differences regarding tumor or treatment-related characteristics between groups. Six of 22 patients (27.3%) in Group 1; 5/13 (38.5%) in Group 2; and 9/13 (69.2%) in Group 3 had cognitive impairment that was considered severe in 3/22 patients (13.6%) in Group 1; 4/13 (30.8%) in Group 2; and 6/13 (46.2%) in Group 3. Patients in Groups 2 and 3 showed significant GM atrophy and more leukoencephalopathy than Group 1. Cognitive deficits were associated with brain atrophy and WM changes. CONCLUSIONS Long-term oligodendroglial tumor survivors who underwent standard RT ± CT treatment, mainly >5 years of its completion, present cognitive impairment, especially on memory and executive functions, associated with late GM and WM damage, thus highlighting the need of developing future strategies in patients with oligodendroglial tumor and long expected survival.
Collapse
Affiliation(s)
- Nuria Cayuela
- Neuro-Oncology Unit, Bellvitge University Hospital, Bellvitge Biomedical Research Institute-Catalan Institute of Oncology (IDIBELL) (Oncobell program), Barcelona, Spain
| | | | - Estela Càmara
- Cognition and Brain Plasticity Group, IDIBELL, Barcelona, Spain
| | - Carles Majós
- Neuro-Oncology Unit, Bellvitge University Hospital, Bellvitge Biomedical Research Institute-Catalan Institute of Oncology (IDIBELL) (Oncobell program), Barcelona, Spain
| | - Noemi Vidal
- Neuro-Oncology Unit, Bellvitge University Hospital, Bellvitge Biomedical Research Institute-Catalan Institute of Oncology (IDIBELL) (Oncobell program), Barcelona, Spain
| | - Anna Lucas
- Neuro-Oncology Unit, Bellvitge University Hospital, Bellvitge Biomedical Research Institute-Catalan Institute of Oncology (IDIBELL) (Oncobell program), Barcelona, Spain
| | - Miguel Gil-Gil
- Neuro-Oncology Unit, Bellvitge University Hospital, Bellvitge Biomedical Research Institute-Catalan Institute of Oncology (IDIBELL) (Oncobell program), Barcelona, Spain
| | - Francesc Graus
- Department of Neurology, August Pi i Sunyer Biomedical Research Institute (IDIBAPS) Hospital Clínic, Barcelona, Spain
| | - Jordi Bruna
- Neuro-Oncology Unit, Bellvitge University Hospital, Bellvitge Biomedical Research Institute-Catalan Institute of Oncology (IDIBELL) (Oncobell program), Barcelona, Spain.,Institute of Neurosciences, Department of Cell Biology, Physiology, and Immunology, Autonomous University of Barcelona, Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), Bellaterra, Spain
| | - Marta Simó
- Neuro-Oncology Unit, Bellvitge University Hospital, Bellvitge Biomedical Research Institute-Catalan Institute of Oncology (IDIBELL) (Oncobell program), Barcelona, Spain.,Cognition and Brain Plasticity Group, IDIBELL, Barcelona, Spain
| |
Collapse
|
220
|
Suckert T, Rassamegevanon T, Müller J, Dietrich A, Graja A, Reiche M, Löck S, Krause M, Beyreuther E, von Neubeck C. Applying Tissue Slice Culture in Cancer Research-Insights from Preclinical Proton Radiotherapy. Cancers (Basel) 2020; 12:E1589. [PMID: 32560230 PMCID: PMC7352770 DOI: 10.3390/cancers12061589] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/16/2020] [Accepted: 06/09/2020] [Indexed: 12/15/2022] Open
Abstract
A challenge in cancer research is the definition of reproducible, reliable, and practical models, which reflect the effects of complex treatment modalities and the heterogeneous response of patients. Proton beam radiotherapy (PBRT), relative to conventional photon-based radiotherapy, offers the potential for iso-effective tumor control, while protecting the normal tissue surrounding the tumor. However, the effects of PBRT on the tumor microenvironment and the interplay with newly developed chemo- and immunotherapeutic approaches are still open for investigation. This work evaluated thin-cut tumor slice cultures (TSC) of head and neck cancer and organotypic brain slice cultures (OBSC) of adult mice brain, regarding their relevance for translational radiooncology research. TSC and OBSC were treated with PBRT and investigated for cell survival with a lactate dehydrogenase (LDH) assay, DNA repair via the DNA double strand break marker γH2AX, as well as histology with regards to morphology. Adult OBSC failed to be an appropriate model for radiobiological research questions. However, histological analysis of TSC showed DNA damage and tumor morphological results, comparable to known in vivo and in vitro data, making them a promising model to study novel treatment approaches in patient-derived xenografts or primary tumor material.
Collapse
Affiliation(s)
- Theresa Suckert
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (T.R.); (A.D.); (A.G.); (S.L.); (M.K.); (C.v.N.)
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01309 Dresden, Germany; (J.M.); (M.R.); (E.B.)
| | - Treewut Rassamegevanon
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (T.R.); (A.D.); (A.G.); (S.L.); (M.K.); (C.v.N.)
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01309 Dresden, Germany; (J.M.); (M.R.); (E.B.)
| | - Johannes Müller
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01309 Dresden, Germany; (J.M.); (M.R.); (E.B.)
- Institute of Radiooncology—OncoRay, Helmholtz-Zentrum Dresden—Rossendorf, 01328 Dresden, Germany
| | - Antje Dietrich
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (T.R.); (A.D.); (A.G.); (S.L.); (M.K.); (C.v.N.)
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01309 Dresden, Germany; (J.M.); (M.R.); (E.B.)
| | - Antonia Graja
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (T.R.); (A.D.); (A.G.); (S.L.); (M.K.); (C.v.N.)
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01309 Dresden, Germany; (J.M.); (M.R.); (E.B.)
| | - Michael Reiche
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01309 Dresden, Germany; (J.M.); (M.R.); (E.B.)
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
| | - Steffen Löck
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (T.R.); (A.D.); (A.G.); (S.L.); (M.K.); (C.v.N.)
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01309 Dresden, Germany; (J.M.); (M.R.); (E.B.)
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01309 Dresden, Germany
| | - Mechthild Krause
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (T.R.); (A.D.); (A.G.); (S.L.); (M.K.); (C.v.N.)
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01309 Dresden, Germany; (J.M.); (M.R.); (E.B.)
- Institute of Radiooncology—OncoRay, Helmholtz-Zentrum Dresden—Rossendorf, 01328 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01309 Dresden, Germany
| | - Elke Beyreuther
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01309 Dresden, Germany; (J.M.); (M.R.); (E.B.)
- Helmholtz-Zentrum Dresden—Rossendorf, Institute of Radiation Physics, 01328 Dresden, Germany
| | - Cläre von Neubeck
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (T.R.); (A.D.); (A.G.); (S.L.); (M.K.); (C.v.N.)
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01309 Dresden, Germany; (J.M.); (M.R.); (E.B.)
- Department of Particle Therapy, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| |
Collapse
|
221
|
Zhang B, Lian Z, Zhong L, Zhang X, Dong Y, Chen Q, Zhang L, Mo X, Huang W, Yang W, Zhang S. Machine-learning based MRI radiomics models for early detection of radiation-induced brain injury in nasopharyngeal carcinoma. BMC Cancer 2020; 20:502. [PMID: 32487085 PMCID: PMC7268644 DOI: 10.1186/s12885-020-06957-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 05/12/2020] [Indexed: 12/15/2022] Open
Abstract
Background Early radiation-induced temporal lobe injury (RTLI) diagnosis in nasopharyngeal carcinoma (NPC) is clinically challenging, and prediction models of RTLI are lacking. Hence, we aimed to develop radiomic models for early detection of RTLI. Methods We retrospectively included a total of 242 NPC patients who underwent regular follow-up magnetic resonance imaging (MRI) examinations, including contrast-enhanced T1-weighted and T2-weighted imaging. For each MRI sequence, four non-texture and 10,320 texture features were extracted from medial temporal lobe, gray matter, and white matter, respectively. The relief and 0.632 + bootstrap algorithms were applied for initial and subsequent feature selection, respectively. Random forest method was used to construct the prediction model. Three models, 1, 2 and 3, were developed for predicting the results of the last three follow-up MRI scans at different times before RTLI onset, respectively. The area under the curve (AUC) was used to evaluate the performance of models. Results Of the 242 patients, 171 (70.7%) were men, and the mean age of all the patients was 48.5 ± 10.4 years. The median follow-up and latency from radiotherapy until RTLI were 46 and 41 months, respectively. In the testing cohort, models 1, 2, and 3, with 20 texture features derived from the medial temporal lobe, yielded mean AUCs of 0.830 (95% CI: 0.823–0.837), 0.773 (95% CI: 0.763–0.782), and 0.716 (95% CI: 0.699–0.733), respectively. Conclusion The three developed radiomic models can dynamically predict RTLI in advance, enabling early detection and allowing clinicians to take preventive measures to stop or slow down the deterioration of RTLI.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China.,Jinan University, Guangzhou, Guangdong, China
| | - Zhouyang Lian
- Department of Radiology, Guangdong Provincial People's Hospital/Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Liming Zhong
- Guangdong Provincial Key Laboratory of Medical Image Processing, School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Xiao Zhang
- Guangdong Provincial Key Laboratory of Medical Image Processing, School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,Zhuhai Precision Medical Center, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Yuhao Dong
- Department of Catheterization Lab, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital /Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, People's Republic of China
| | - Qiuying Chen
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China.,Jinan University, Guangzhou, Guangdong, China
| | - Lu Zhang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China.,Jinan University, Guangzhou, Guangdong, China
| | - Xiaokai Mo
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Wenhui Huang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Wei Yang
- Guangdong Provincial Key Laboratory of Medical Image Processing, School of Biomedical Engineering, Southern Medical University, Guangzhou, China.
| | - Shuixing Zhang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China.
| |
Collapse
|
222
|
Kocher M, Jockwitz C, Caspers S, Schreiber J, Farrher E, Stoffels G, Filss C, Lohmann P, Tscherpel C, Ruge MI, Fink GR, Shah NJ, Galldiks N, Langen KJ. Role of the default mode resting-state network for cognitive functioning in malignant glioma patients following multimodal treatment. Neuroimage Clin 2020; 27:102287. [PMID: 32540630 PMCID: PMC7298724 DOI: 10.1016/j.nicl.2020.102287] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/31/2020] [Accepted: 04/27/2020] [Indexed: 01/03/2023]
Abstract
BACKGROUND Progressive cognitive decline following multimodal neurooncological treatment is a common observation in patients suffering from malignant glioma. Alterations of the default-mode network (DMN) represent a possible source of impaired neurocognitive functioning and were analyzed in these patients. METHODS Eighty patients (median age, 51 years) with glioma (WHO grade IV glioblastoma, n = 57; WHO grade III anaplastic astrocytoma, n = 13; WHO grade III anaplastic oligodendroglioma, n = 10) and ECOG performance score 0-1 underwent resting-state functional MRI (rs-fMRI) and neuropsychological testing at a median interval of 13 months (range, 1-114 months) after initiation of therapy. For evaluation of structural and metabolic changes after treatment, anatomical MRI and amino acid PET using O-(2-[18F]fluoroethyl)-L-tyrosine (FET) were simultaneously acquired to rs-fMRI on a hybrid MR/PET scanner. A cohort of 80 healthy subjects matched for gender, age, and educational status served as controls. RESULTS The connectivity pattern within the DMN (12 nodes) of the glioma patients differed significantly from that of the healthy subjects but did not depend on age, tumor grade, time since treatment initiation, presence of residual/recurrent tumor, number of chemotherapy cycles received, or anticonvulsive medication. Small changes in the connectivity pattern were observed in patients who had more than one series of radiotherapy. In contrast, structural tissue changes located at or near the tumor site (including resection cavities, white matter lesions, edema, and tumor tissue) had a strong negative impact on the functional connectivity of the adjacent DMN nodes, resulting in a marked dependence of the connectivity pattern on tumor location. In the majority of neurocognitive domains, glioma patients performed significantly worse than healthy subjects. Correlation analysis revealed that reduced connectivity in the left temporal and parietal DMN nodes was associated with low performance in language processing and verbal working memory. Furthermore, connectivity of the left parietal DMN node also correlated with processing speed, executive function, and verbal as well as visual working memory. Overall DMN connectivity loss and cognitive decline were less pronounced in patients with higher education. CONCLUSION Personalized treatment strategies for malignant glioma patients should consider the left parietal and temporal DMN nodes as vulnerable regions concerning neurocognitive outcome.
Collapse
Affiliation(s)
- Martin Kocher
- Institute of Neuroscience and Medicine (INM-1, -3, -4), Research Center Juelich, Wilhelm-Johnen-Str., 52428 Juelich, Germany; Department of Stereotaxy and Functional Neurosurgery, Center for Neurosurgery, Faculty of Medicine and University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany; Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne and Duesseldorf, Kerpener Str. 62, 50937 Cologne, Germany.
| | - Christiane Jockwitz
- Institute of Neuroscience and Medicine (INM-1, -3, -4), Research Center Juelich, Wilhelm-Johnen-Str., 52428 Juelich, Germany; Department of Psychiatry, Psychotherapy and Psychosomatics, RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, Germany
| | - Svenja Caspers
- Institute of Neuroscience and Medicine (INM-1, -3, -4), Research Center Juelich, Wilhelm-Johnen-Str., 52428 Juelich, Germany; Juelich-Aachen Research Alliance (JARA)-Section JARA-Brain, Wilhelm-Johnen-Str., 52428 Juelich, Germany; Institute for Anatomy I, Medical Faculty, Heinrich Heine University Duesseldorf, Universitaetsstr. 1, 40225 Duesseldorf, Germany
| | - Jan Schreiber
- Institute of Neuroscience and Medicine (INM-1, -3, -4), Research Center Juelich, Wilhelm-Johnen-Str., 52428 Juelich, Germany
| | - Ezequiel Farrher
- Institute of Neuroscience and Medicine (INM-1, -3, -4), Research Center Juelich, Wilhelm-Johnen-Str., 52428 Juelich, Germany
| | - Gabriele Stoffels
- Institute of Neuroscience and Medicine (INM-1, -3, -4), Research Center Juelich, Wilhelm-Johnen-Str., 52428 Juelich, Germany
| | - Christian Filss
- Institute of Neuroscience and Medicine (INM-1, -3, -4), Research Center Juelich, Wilhelm-Johnen-Str., 52428 Juelich, Germany
| | - Philipp Lohmann
- Institute of Neuroscience and Medicine (INM-1, -3, -4), Research Center Juelich, Wilhelm-Johnen-Str., 52428 Juelich, Germany; Department of Stereotaxy and Functional Neurosurgery, Center for Neurosurgery, Faculty of Medicine and University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Caroline Tscherpel
- Institute of Neuroscience and Medicine (INM-1, -3, -4), Research Center Juelich, Wilhelm-Johnen-Str., 52428 Juelich, Germany; Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne and Duesseldorf, Kerpener Str. 62, 50937 Cologne, Germany
| | - Maximilian I Ruge
- Department of Stereotaxy and Functional Neurosurgery, Center for Neurosurgery, Faculty of Medicine and University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany; Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne and Duesseldorf, Kerpener Str. 62, 50937 Cologne, Germany
| | - Gereon R Fink
- Institute of Neuroscience and Medicine (INM-1, -3, -4), Research Center Juelich, Wilhelm-Johnen-Str., 52428 Juelich, Germany; Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Nadim J Shah
- Institute of Neuroscience and Medicine (INM-1, -3, -4), Research Center Juelich, Wilhelm-Johnen-Str., 52428 Juelich, Germany; Institute of Neuroscience and Medicine 11, JARA, Research Center Juelich, Wilhelm-Johnen-Str., 52428 Juelich, Germany; Juelich-Aachen Research Alliance (JARA)-Section JARA-Brain, Wilhelm-Johnen-Str., 52428 Juelich, Germany; Department of Neurology, University Hospital Aachen, RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, Germany
| | - Norbert Galldiks
- Institute of Neuroscience and Medicine (INM-1, -3, -4), Research Center Juelich, Wilhelm-Johnen-Str., 52428 Juelich, Germany; Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany; Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne and Duesseldorf, Kerpener Str. 62, 50937 Cologne, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-1, -3, -4), Research Center Juelich, Wilhelm-Johnen-Str., 52428 Juelich, Germany; Department of Nuclear Medicine, University Hospital Aachen, RWTH Aachen University, Pauwelsstr. 30, 52074 Aachen, Germany
| |
Collapse
|
223
|
Clément GR, Boyle RD, George KA, Nelson GA, Reschke MF, Williams TJ, Paloski WH. Challenges to the central nervous system during human spaceflight missions to Mars. J Neurophysiol 2020; 123:2037-2063. [DOI: 10.1152/jn.00476.2019] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Space travel presents a number of environmental challenges to the central nervous system, including changes in gravitational acceleration that alter the terrestrial synergies between perception and action, galactic cosmic radiation that can damage sensitive neurons and structures, and multiple factors (isolation, confinement, altered atmosphere, and mission parameters, including distance from Earth) that can affect cognition and behavior. Travelers to Mars will be exposed to these environmental challenges for up to 3 years, and space-faring nations continue to direct vigorous research investments to help elucidate and mitigate the consequences of these long-duration exposures. This article reviews the findings of more than 50 years of space-related neuroscience research on humans and animals exposed to spaceflight or analogs of spaceflight environments, and projects the implications and the forward work necessary to ensure successful Mars missions. It also reviews fundamental neurophysiology responses that will help us understand and maintain human health and performance on Earth.
Collapse
Affiliation(s)
| | - Richard D. Boyle
- National Aeronautics and Space Administration, Ames Research Center, Moffett Field, California
| | | | - Gregory A. Nelson
- Division of Biomedical Engineering Sciences, School of Medicine Loma Linda University, Loma Linda, California
| | - Millard F. Reschke
- National Aeronautics and Space Administration, Johnson Space Center, Houston, Texas
| | - Thomas J. Williams
- National Aeronautics and Space Administration, Johnson Space Center, Houston, Texas
| | - William H. Paloski
- National Aeronautics and Space Administration, Johnson Space Center, Houston, Texas
| |
Collapse
|
224
|
Turnquist C, Beck JA, Horikawa I, Obiorah IE, Von Muhlinen N, Vojtesek B, Lane DP, Grunseich C, Chahine JJ, Ames HM, Smart DD, Harris BT, Harris CC. Radiation-induced astrocyte senescence is rescued by Δ133p53. Neuro Oncol 2020; 21:474-485. [PMID: 30615147 DOI: 10.1093/neuonc/noz001] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Cellular senescence and the senescence-associated secretory phenotype (SASP) may contribute to the development of radiation therapy-associated side effects in the lung and blood vessels by promoting chronic inflammation. In the brain, inflammation contributes to the development of neurologic disease, including Alzheimer's disease. In this study, we investigated the roles of cellular senescence and Δ133p53, an inhibitory isoform of p53, in radiation-induced brain injury. METHODS Senescent cell types in irradiated human brain were identified with immunohistochemical labeling of senescence-associated proteins p16INK4A and heterochromatin protein Hp1γ in 13 patient cases, including 7 irradiated samples. To investigate the impact of radiation on astrocytes specifically, primary human astrocytes were irradiated and examined for expression of Δ133p53 and induction of SASP. Lentiviral expression of ∆133p53 was performed to investigate its role in regulating radiation-induced cellular senescence and astrocyte-mediated neuroinflammation. RESULTS Astrocytes expressing p16INK4A and Hp1γ were identified in all irradiated tissues, were increased in number in irradiated compared with untreated cancer patient tissues, and had higher labeling intensity in irradiated tissues compared with age-matched controls. Human astrocytes irradiated in vitro also experience induction of cellular senescence, have diminished Δ133p53, and adopt a neurotoxic phenotype as demonstrated by increased senescence-associated beta-galactosidase activity, p16INK4A, and interleukin (IL)-6. In human astrocytes, Δ133p53 inhibits radiation-induced senescence, promotes DNA double-strand break repair, and prevents astrocyte-mediated neuroinflammation and neurotoxicity. CONCLUSIONS Restoring expression of the endogenous p53 isoform, ∆133p53, protects astrocytes from radiation-induced senescence, promotes DNA repair, and inhibits astrocyte-mediated neuroinflammation.
Collapse
Affiliation(s)
- Casmir Turnquist
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jessica A Beck
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Izumi Horikawa
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ifeyinwa E Obiorah
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
| | - Natalia Von Muhlinen
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Borivoj Vojtesek
- Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - David P Lane
- p53 Laboratory, Biomedical Sciences Institutes (A*STAR), Singapore
| | - Christopher Grunseich
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Joeffrey J Chahine
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA
| | - Heather M Ames
- Department of Pathology, Johns Hopkins Hospital, Baltimore, Maryland, USA.,Department of Pathology, University of Maryland, Baltimore, Maryland, USA
| | - Dee Dee Smart
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Brent T Harris
- Department of Pathology, Georgetown University Medical Center, Washington, DC, USA.,Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
225
|
Hughes RT, McTyre ER, LeCompte M, Cramer CK, Munley MT, Laxton AW, Tatter SB, Ruiz J, Pasche B, Watabe K, Chan MD. Clinical Outcomes of Upfront Stereotactic Radiosurgery Alone for Patients With 5 to 15 Brain Metastases. Neurosurgery 2020; 85:257-263. [PMID: 29982831 DOI: 10.1093/neuros/nyy276] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 05/30/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND The role of primary stereotactic radiosurgery (SRS) for patients with >4 brain metastases (BM) remains controversial. OBJECTIVE To compare the outcomes of patients treated with upfront SRS alone for 1, 2 to 4, and 5 to 15 BM and assess for predictors of clinical outcomes in the 5 to 15 BM group. METHODS A total of 478 patients treated with upfront SRS were stratified by number of lesions: 220 had 1 BM, 190 had 2 to 4 BM, and 68 patients had 5 to 15 BM. Overall survival and whole brain radiotherapy-free survival were estimated using the Kaplan-Meier method. The cumulative incidences of local failure and distant brain failure (DBF) were estimated using competing risks methodology. Clinicopathologic and dosimetric parameters were evaluated as predictors of survival and DBF in patients with 5 to 15 BM using Cox proportional hazards. RESULTS Median overall survival was 8.0, 6.3, and 4.7 mo for patients with 1, 2 to 4, and 5 to 15 BM, respectively (P = .14). One-year DBF was 27%, 44%, and 40%, respectively (P = .01). Salvage SRS and whole brain radiotherapy rates did not differ. Progressive extracranial disease and gastrointestinal primary were associated with poor survival while RCC primary was associated with increased risk of DBF. No evaluated dose-volume parameters predicted for death, neurologic death or toxicity. CONCLUSION SRS for 5 to 15 BM is well tolerated without evidence of an associated increase in toxicity, treatment failure, or salvage therapy. Further prospective, randomized studies are warranted to clarify the role of SRS for these patients.
Collapse
Affiliation(s)
- Ryan T Hughes
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Emory R McTyre
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Michael LeCompte
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Christina K Cramer
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Michael T Munley
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Adrian W Laxton
- Department of Neurosurgery, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Stephen B Tatter
- Department of Neurosurgery, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Jimmy Ruiz
- Department of Medicine (Hematology & Oncology), Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Boris Pasche
- Department of Medicine (Hematology & Oncology), Wake Forest School of Medicine, Winston Salem, North Carolina.,Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Kounosuke Watabe
- Department of Cancer Biology, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Michael D Chan
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston Salem, North Carolina
| |
Collapse
|
226
|
Kim SB, Heo JI, Kim H, Kim KS. Acetylation of PGC1α by Histone Deacetylase 1 Downregulation Is Implicated in Radiation-Induced Senescence of Brain Endothelial Cells. J Gerontol A Biol Sci Med Sci 2020; 74:787-793. [PMID: 30016403 DOI: 10.1093/gerona/gly167] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Indexed: 12/21/2022] Open
Abstract
Peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC1α) is a potent transcription factor for mitochondrial function, lipid metabolism, and detoxification in a variety of tissues. PGC1α also promotes brain cell proliferation and memory. However, how PGC1α is involved in aging is not well known. In brain endothelial cells, we found that PGC1α knockdown accelerated DNA damage-induced senescence, evidenced by an increase in senescence-associated β-galactosidase-positive cells and a decrease in cell proliferation and adenosine triphosphate production. PGC1α knockdown delayed DNA damage repair mechanisms compared with the wild-type condition as shown by γ-H2AX foci staining assay. Overexpression of PGC1α reduced senescence-associated β-galactosidase-positive cells and increased the proliferation of senescent cells. Although PGC1α protein levels were not decreased, PGC1 acetylation was increased by ionizing radiation treatment and aging. Histone deacetylase 1 (HDAC1) expression was decreased by ionizing radiation treatment and aging, and downregulation of HDAC1 induced acetylation of PGC1α. HDAC1 knockdown affected sirtuin 1 expression and decreased its deacetylation of PGC1α. In the mouse brain cortex, acetylation of PGC1α was increased by ionizing radiation treatment. These results suggest that acetylation of PGC1α is induced by DNA damage agents such as ionizing radiation, which deregulates mitochondrial mechanisms and metabolism, resulting in acceleration of radiation-induced senescence. Therefore, acetylation of PGC1α may be a cause of brain disorders and has the potential to serve as a therapeutic target for radiation-induced senescence after radiation cancer therapy.
Collapse
Affiliation(s)
- Su-Bin Kim
- Division of Applied Radiation Bioscience, Korea Institute of Radiological and Medical Sciences, Seoul
- Department of Biotechnology, School of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Jong-Ik Heo
- Division of Applied Radiation Bioscience, Korea Institute of Radiological and Medical Sciences, Seoul
| | - Hyunggee Kim
- Department of Biotechnology, School of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Kwang Seok Kim
- Division of Applied Radiation Bioscience, Korea Institute of Radiological and Medical Sciences, Seoul
| |
Collapse
|
227
|
Ivanov VN, Grabham PW, Wu CC, Hei TK. Inhibition of autophagic flux differently modulates cannabidiol-induced death in 2D and 3D glioblastoma cell cultures. Sci Rep 2020; 10:2687. [PMID: 32060308 PMCID: PMC7021896 DOI: 10.1038/s41598-020-59468-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023] Open
Abstract
Radiotherapy combined with chemotherapy is the major treatment modality for human glioblastoma multiforme (GBM). GBMs eventually relapse after treatment and the average survival of GBM patients is less than two years. There is some evidence that cannabidiol (CBD) can induce cell death and increases the radiosensitivity of GBM by enhancing apoptosis. Beside initiation of death, CBD has been demonstrated as an inducer of autophagy. In the present study, we address the question whether CBD simultaneously induces a protective effect in GBM by upregulating autophagy. Addition of chloroquine that suppressed autophagic flux to 2D GBM cultures increased CBD-induced cell death, presenting proof for the protective autophagy. Blockage of autophagy upregulated radiation-induced cytotoxicity but only modestly affected the levels of cell death in CBD- or CBD/γ-irradiated 3D GBM cultures. Furthermore, CBD enhanced the pro-apoptotic activities of JNK1/2 and MAPK p38 signaling cascades while partially downregulated the pro-survival PI3K-AKT cascade, thereby changing a balance between cell death and survival. Suppression of JNK activation partially reduced CBD-induced cell death in 3D GBM cultures. In contrast, co-treatment of CBD-targeted cells with inhibitors of PI3K-AKT-NF-κB, IKK-NF-κB or JAK2-STAT3 pathways killed surviving GBM cells in both 2D and 3D cultures, potentially improving the therapeutic ratio of GBM.
Collapse
Affiliation(s)
- Vladimir N Ivanov
- Center for Radiological Research, Department of Radiation Oncology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA.
| | - Peter W Grabham
- Center for Radiological Research, Department of Radiation Oncology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Cheng-Chia Wu
- Center for Radiological Research, Department of Radiation Oncology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - Tom K Hei
- Center for Radiological Research, Department of Radiation Oncology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| |
Collapse
|
228
|
Remes TM, Suo-Palosaari MH, Koskenkorva PKT, Sutela AK, Toiviainen-Salo SM, Arikoski PM, Arola MO, Heikkilä VP, Kapanen M, Lähteenmäki PM, Lönnqvist TRI, Niiniviita H, Pokka TML, Porra L, Riikonen VP, Seppälä J, Sirkiä KH, Vanhanen A, Rantala HMJ, Harila-Saari AH, Ojaniemi MK. Radiation-induced accelerated aging of the brain vasculature in young adult survivors of childhood brain tumors. Neurooncol Pract 2020; 7:415-427. [PMID: 32760593 PMCID: PMC7393284 DOI: 10.1093/nop/npaa002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Background Cranial radiotherapy may damage the cerebral vasculature. The aim of this study was to understand the prevalence and risk factors of cerebrovascular disease (CVD) and white matter hyperintensities (WMHs) in childhood brain tumors (CBT) survivors treated with radiotherapy. Methods Seventy CBT survivors who received radiotherapy were enrolled in a cross-sectional study at a median 20 years after radiotherapy cessation. The prevalence of and risk factors for CVD were investigated using MRI, MRA, and laboratory testing. Tumors, their treatment, and stroke-related data were retrieved from patients’ files. Results Forty-four individuals (63%) had CVD at a median age of 27 years (range, 16-43 years). The prevalence rates at 20 years for CVD, small-vessel disease, and large-vessel disease were 52%, 38%, and 16%, respectively. Ischemic infarcts were diagnosed in 6 survivors, and cerebral hemorrhage in 2. Lacunar infarcts were present in 7, periventricular or deep WMHs in 34 (49%), and mineralizing microangiopathy in 21 (30%) survivors. Multiple pathologies were detected in 44% of the participants, and most lesions were located in a high-dose radiation area. Higher blood pressure was associated with CVD and a presence of WMHs. Higher cholesterol levels increased the risk of ischemic infarcts and WMHs, and lower levels of high-density lipoprotein and higher waist circumference increased the risk of lacunar infarcts. Conclusions Treating CBTs with radiotherapy increases the risk of early CVD and WMHs in young adult survivors. These results suggest an urgent need for investigating CVD prevention in CBT patients.
Collapse
Affiliation(s)
- Tiina Maria Remes
- Department of Pediatrics and Adolescence, PEDEGO Research Unit and Medical Research Center, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Maria Helena Suo-Palosaari
- Department of Diagnostic Radiology, Oulu University Hospital, and University of Oulu, Research Unit of Medical Imaging, Physics, and Technology, Faculty of Medicine, University of Oulu, and Medical Research Center Oulu, University of Oulu, Oulu, Finland
| | | | - Anna K Sutela
- Department of Clinical Radiology, Kuopio University Hospital, Kuopio, Finland
| | - Sanna-Maria Toiviainen-Salo
- Department of Pediatric Radiology, HUS Medical Imaging Center, Radiology, University of Helsinki, and Helsinki University Hospital, Helsinki, Finland
| | - Pekka M Arikoski
- Department of Pediatrics and Adolescence, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
| | - Mikko O Arola
- Department of Pediatrics, Tampere University Hospital, and University of Tampere, Tampere, Finland
| | - Vesa-Pekka Heikkilä
- Department of Oncology and Radiotherapy, Oulu University Hospital, Oulu, Finland
| | - Mika Kapanen
- Department of Oncology and Department of Medical Physics, Tampere University Hospital, Tampere, Finland
| | - Päivi Maria Lähteenmäki
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital, and Turku University, Turku, Finland
| | - Tuula R I Lönnqvist
- Department of Child Neurology, Children's Hospital, University of Helsinki, and Helsinki University Hospital, Helsinki, Finland
| | - Hannele Niiniviita
- Department of Medical Physics, Division of Medical Imaging, Turku University Hospital, Turku, Finland
| | - Tytti M-L Pokka
- Department of Pediatrics and Adolescence, PEDEGO Research Unit and Medical Research Center, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Liisa Porra
- Department of Oncology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - V Pekka Riikonen
- Department of Pediatrics and Adolescence, Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland
| | - Jan Seppälä
- Center of Oncology, Kuopio University Hospital, Kuopio, Finland
| | - Kirsti H Sirkiä
- Department of Pediatrics and Adolescence, Helsinki University, and Helsinki University Hospital, Helsinki, Finland
| | - Antti Vanhanen
- Department of Oncology and Department of Medical Physics, Tampere University Hospital, Tampere, Finland
| | - Heikki M J Rantala
- Department of Pediatrics and Adolescence, PEDEGO Research Unit and Medical Research Center, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Arja H Harila-Saari
- Uppsala University, Department of Women's and Children's Health, Akademiska sjukhuset, Uppsala, Sweden
| | - Marja K Ojaniemi
- Department of Pediatrics and Adolescence, PEDEGO Research Unit and Medical Research Center, Oulu University Hospital, and University of Oulu, Oulu, Finland
| |
Collapse
|
229
|
Verhaak E, Gehring K, Hanssens PEJ, Aaronson NK, Sitskoorn MM. Health-related quality of life in adult patients with brain metastases after stereotactic radiosurgery: a systematic, narrative review. Support Care Cancer 2020; 28:473-484. [PMID: 31792879 PMCID: PMC6954134 DOI: 10.1007/s00520-019-05136-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 10/16/2019] [Indexed: 01/03/2023]
Abstract
PURPOSE A growing number of patients with brain metastases (BM) are being treated with stereotactic radiosurgery (SRS), and the importance of evaluating the impact of SRS on the health-related quality of life (HRQoL) in these patients has been increasingly acknowledged. This systematic review summarizes the current knowledge about the HRQoL of patients with BM after SRS. METHODS We searched EMBASE, Medline Ovid, Web-of-Science, the Cochrane Database, PsycINFO Ovid, and Google Scholar up to November 15, 2018. Studies in patients with BM in which HRQoL was assessed before and after SRS and analyzed over time were included. Studies including populations of several types of brain cancer and/or several types of treatments were included if the results for patients with BM and treatment with SRS alone were described separately. RESULTS Out of 3638 published articles, 9 studies met the eligibility criteria and were included. In 4 out of 7 studies on group results, overall HRQoL of patients with BM remained stable after SRS. In small study samples of longer-term survivors, overall HRQoL remained stable up to 12 months post-SRS. Contradictory results were reported for physical and general/global HRQoL, which might be explained by the different questionnaires that were used. CONCLUSIONS In general, SRS does not have significant negative effects on patients' overall HRQoL over time. Future research is needed to analyze different aspects of HRQoL, differences in individual changes in HRQoL after SRS, and factors that influence these changes. These studies should take into account several methodological issues as discussed in this review.
Collapse
Affiliation(s)
- Eline Verhaak
- Gamma Knife Center, Elisabeth-TweeSteden Hospital, Hilvarenbeekseweg 60, 5022 GC, Tilburg, The Netherlands
- Department of Neurosurgery, Elisabeth-TweeSteden Hospital, Hilvarenbeekseweg 60, 5022 GC, Tilburg, The Netherlands
- Department of Cognitive Neuropsychology, Tilburg University, Warandelaan 2, 5037 AB, Tilburg, The Netherlands
| | - Karin Gehring
- Department of Neurosurgery, Elisabeth-TweeSteden Hospital, Hilvarenbeekseweg 60, 5022 GC, Tilburg, The Netherlands.
- Department of Cognitive Neuropsychology, Tilburg University, Warandelaan 2, 5037 AB, Tilburg, The Netherlands.
| | - Patrick E J Hanssens
- Gamma Knife Center, Elisabeth-TweeSteden Hospital, Hilvarenbeekseweg 60, 5022 GC, Tilburg, The Netherlands
- Department of Neurosurgery, Elisabeth-TweeSteden Hospital, Hilvarenbeekseweg 60, 5022 GC, Tilburg, The Netherlands
| | - Neil K Aaronson
- Division of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Margriet M Sitskoorn
- Department of Neurosurgery, Elisabeth-TweeSteden Hospital, Hilvarenbeekseweg 60, 5022 GC, Tilburg, The Netherlands
- Department of Cognitive Neuropsychology, Tilburg University, Warandelaan 2, 5037 AB, Tilburg, The Netherlands
| |
Collapse
|
230
|
Cerebral microbleeds in adult survivors of childhood acute lymphoblastic leukemia treated with cranial radiation. Sci Rep 2020; 10:692. [PMID: 31959839 PMCID: PMC6971068 DOI: 10.1038/s41598-020-57682-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 01/03/2020] [Indexed: 01/14/2023] Open
Abstract
Cranial radiation therapy is associated with white matter-specific brain injury, cortical volume loss, mineralization, microangiopathy and neurocognitive impairment in survivors of childhood acute lymphoblastic leukemia. In this retrospective cross-sectional analysis, neurocognitive testing and 3 T brain MRI’s were obtained in 101 survivors treated with cranial radiation. Small focal intracerebral hemorrhages only visible on exquisitely sensitive MRI sequences were identified and localized using susceptibility weighted imaging. Modified Poisson regression was used to assess the effect of cranial radiation on cumulative number and location of microbleeds in each brain region, and multiple linear regression was used to evaluate microbleeds on neurocognitive outcomes, adjusting for age at diagnosis and sex. At least one microbleed was present in 85% of survivors, occurring more frequently in frontal lobes. Radiation dose of 24 Gy conveyed a 5-fold greater risk (95% CI 2.57–10.32) of having multiple microbleeds compared to a dose of 18 Gy. No significant difference was found in neurocognitive scores with either the absence or presence of microbleeds or their location. Greater prevalence of microbleeds in our study compared to prior reports is likely related to longer time since treatment, better sensitivity of SWI for detection of microbleeds and the use of a 3 T MRI platform.
Collapse
|
231
|
Endothelial Autophagy: an Effective Target for Radiation-induced Cerebral Capillary Damage. Sci Rep 2020; 10:614. [PMID: 31953486 PMCID: PMC6968992 DOI: 10.1038/s41598-019-57234-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 12/11/2019] [Indexed: 02/05/2023] Open
Abstract
Toxicity to central nervous system tissues is the common side effects for radiotherapy of brain tumor. The radiation toxicity has been thought to be related to the damage of cerebral endothelium. However, because of lacking a suitable high-resolution vivo model, cellular response of cerebral capillaries to radiation remained unclear. Here, we present the flk:eGFP transgenic zebrafish larvae as a feasible model to study the radiation toxicity to cerebral capillary. We showed that, in living zebrafish larvae, radiation could induce acute cerebral capillary shrinkage and blood-flow obstruction, resulting brain hypoxia and glycolysis retardant. Although in vivo neuron damage was also observed after the radiation exposure, further investigation found that they didn’t response to the same dosage of radiation in vitro, indicating that radiation induced neuron damage was a secondary-effect of cerebral vascular function damage. In addition, transgenic labeling and qPCR results showed that the radiation-induced acute cerebral endothelial damage was correlated with intensive endothelial autophagy. Different autophagy inhibitors could significantly alleviate the radiation-induced cerebral capillary damage and prolong the survival of zebrafish larvae. Therefore, we showed that radiation could directly damage cerebral capillary, resulting to blood flow deficiency and neuron death, which suggested endothelial autophagy as a potential target for radiation-induced brain toxicity.
Collapse
|
232
|
Lv X, He H, Yang Y, Han L, Guo Z, Chen H, Li J, Qiu Y, Xie C. Radiation-induced hippocampal atrophy in patients with nasopharyngeal carcinoma early after radiotherapy: a longitudinal MR-based hippocampal subfield analysis. Brain Imaging Behav 2020; 13:1160-1171. [PMID: 30054872 DOI: 10.1007/s11682-018-9931-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Increasing evidence indicates that radiation-induced injury to the hippocampus may play a critical role in neurocognitive dysfunction in patients with nasopharyngeal carcinoma (NPC). However, few studies have assessed RT-induced hippocampal structural alterations in these patients early after radiotherapy (RT). In this study, 58 NPC patients were longitudinally followed up prior to treatment initiation as well as 3 and 6 months after RT, respectively. Twenty comparable normal controls were recruited and followed up in parallel. A novel magnetic resonance imaging (MRI)-based automated method was used to label hippocampal subfields. The linear mixed model was employed to evaluate longitudinal changes in the volumes of the whole hippocampus and seven hippocampal subfields. Time-dependent volume reduction was observed in the bilateral hippocampus, as well as in the bilateral granule cell layer (GCL), bilateral cornu ammonis 1 (CA1), bilateral molecular layer (ML), and bilateral subiculum (SUB) in NPC patients, but not in controls. Moreover, volume deficits in the bilateral hippocampus, bilateral GCL, and right ML showed dose-dependent patterns, and high volume losses in the bilateral hippocampus, bilateral GCL, left SUB, and right ML were associated with a rapid decline in cognitive function. Our findings demonstrated that the hippocampal subfields were selectively injured by irradiation-related early neurotoxic effects, which might account for cognitive impairment in NPC patients at an early stage after RT. Further, structural MRI could serve as a potential noninvasive imaging biomarker for the early detection of radiation effects on the hippocampus in NPC patients after RT.
Collapse
Affiliation(s)
- Xiaofei Lv
- Department of Medical Imaging, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, People's Republic of China
| | - Haoqiang He
- Department of Medical Imaging, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, People's Republic of China
| | - Yadi Yang
- Department of Medical Imaging, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, People's Republic of China
| | - Lujun Han
- Department of Medical Imaging, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, People's Republic of China
| | - Zheng Guo
- Department of Oncology, The First Affiliated Hospital of Ganzhou Medical University, Ganzhou, Jiangxi, People's Republic of China
| | - Hong Chen
- Department of Medical Imaging, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, People's Republic of China
| | - Jing Li
- Department of Medical Imaging, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, People's Republic of China
| | - Yingwei Qiu
- Department of Radiology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, People's Republic of China.
| | - Chuanmiao Xie
- Department of Medical Imaging, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, People's Republic of China.
| |
Collapse
|
233
|
Monti S, Paganelli C, Buizza G, Preda L, Valvo F, Baroni G, Palma G, Cella L. A novel framework for spatial normalization of dose distributions in voxel-based analyses of brain irradiation outcomes. Phys Med 2020; 69:164-169. [PMID: 31918368 DOI: 10.1016/j.ejmp.2019.12.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 12/05/2019] [Accepted: 12/17/2019] [Indexed: 11/29/2022] Open
Abstract
PURPOSE To devise a novel Spatial Normalization framework for Voxel-based analysis (VBA) in brain radiotherapy. VBAs rely on accurate spatial normalization of different patients' planning CTs on a common coordinate system (CCS). The cerebral anatomy, well characterized by MRI, shows instead poor contrast in CT, resulting in potential inaccuracies in VBAs based on CT alone. METHODS We analyzed 50 meningioma patients treated with proton-therapy, undergoing planning CT and T1-weighted (T1w) MRI. The spatial normalization pipeline based on MR and CT images consisted in: intra-patient registration of CT to T1w, inter-patient registration of T1w to MNI space chosen as CCS, doses propagation to MNI. The registration quality was compared with that obtained by Statistical Parametric Mapping software (SPM), used as benchmark. To evaluate the accuracy of dose normalization, the dose organ overlap (DOO) score was computed on gray matter, white matter and cerebrospinal fluid before and after normalization. In addition, the trends in the DOOs distribution were investigated by means of cluster analysis. RESULTS The registration quality was higher for the proposed method compared to SPM (p < 0.001). The DOO scores showed a significant improvement after normalization (p < 0.001). The cluster analysis highlighted 2 clusters, with one of them including the majority of data and exhibiting acceptable DOOs. CONCLUSIONS Our study presents a robust tool for spatial normalization, specifically tailored for brain dose VBAs. Furthermore, the cluster analysis provides a formal criterion for patient exclusion in case of non-acceptable normalization results. The implemented framework lays the groundwork for future reliable VBAs in brain irradiation studies.
Collapse
Affiliation(s)
- S Monti
- National Research Council, Institute of Biostructures and Bioimaging, Napoli, Italy.
| | - C Paganelli
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - G Buizza
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - L Preda
- National Centre of Oncological Hadrontherapy, Pavia, Italy
| | - F Valvo
- National Centre of Oncological Hadrontherapy, Pavia, Italy
| | - G Baroni
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - G Palma
- National Research Council, Institute of Biostructures and Bioimaging, Napoli, Italy
| | - L Cella
- National Research Council, Institute of Biostructures and Bioimaging, Napoli, Italy
| |
Collapse
|
234
|
Roy SJ, Koval OM, Sebag SC, Ait-Aissa K, Allen BG, Spitz DR, Grumbach IM. Inhibition of CaMKII in mitochondria preserves endothelial barrier function after irradiation. Free Radic Biol Med 2020; 146:287-298. [PMID: 31711984 PMCID: PMC7274136 DOI: 10.1016/j.freeradbiomed.2019.11.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 01/03/2023]
Abstract
Damage to the microvascular endothelium is an important part of normal tissue injury after radiation exposure and driven by the production of pro-oxidants. The Ca2+/calmodulin-dependent protein kinase II is present in the mitochondrial matrix (mitoCaMKII) where it regulates Ca2+ uptake via the mitochondrial Ca2+ uniporter (MCU) and pro-oxidant production. Here, we demonstrate that radiation exposure disrupts endothelial cell barrier integrity in vitro, but can be abrogated by inhibition of mitoCaMKII, MCU, or opening of the mitochondrial transition pore. Scavenging of mitochondrial pro-oxidants with mitoTEMPO before, but not after irradiation, protected barrier function. Furthermore, markers of apoptosis and mitochondrial pro-oxidant production were elevated at 24 h following irradiation and abolished by mitoCaMKII inhibition. Endothelial barrier dysfunction was detected as early as 2 h after irradiation. Despite only mildly impaired mitochondrial respiration, the intracellular ATP levels were significantly reduced 4 h after irradiation and correlated with barrier function. MitoCaMKII inhibition improved intracellular ATP concentrations by increasing glycolysis. Finally, DNA double strand break repair and non-homologous end joining, two major drivers of ATP consumption after irradiation, were greatly increased but not significantly affected by mitoCaMKII inhibition. These findings support the hypothesis that mitoCaMKII activity is linked to mitochondrial pro-oxidant production, reduced ATP production, and loss of endothelial barrier function following irradiation. The inhibition of mitoCaMKII is a promising approach to limiting radiation-induced endothelial injury.
Collapse
Affiliation(s)
- Stephen J Roy
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, 169 Newton Rd, 4336 PBDB, University of Iowa, Iowa City, IA, 52242, USA
| | - Olha M Koval
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, 169 Newton Rd, 4336 PBDB, University of Iowa, Iowa City, IA, 52242, USA
| | - Sara C Sebag
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, 169 Newton Rd, 4336 PBDB, University of Iowa, Iowa City, IA, 52242, USA
| | - Karima Ait-Aissa
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, 169 Newton Rd, 4336 PBDB, University of Iowa, Iowa City, IA, 52242, USA
| | - Bryan G Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, B180 Medical Laboratories, University of Iowa, Iowa City, IA, 52242, USA
| | - Douglas R Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, B180 Medical Laboratories, University of Iowa, Iowa City, IA, 52242, USA
| | - Isabella M Grumbach
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, 169 Newton Rd, 4336 PBDB, University of Iowa, Iowa City, IA, 52242, USA; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, B180 Medical Laboratories, University of Iowa, Iowa City, IA, 52242, USA; Veterans Affairs Health Care System, 601 Hwy 6 West Iowa City, IA, 52246, USA.
| |
Collapse
|
235
|
Smith SM, Giedzinski E, Angulo MC, Lui T, Lu C, Park AL, Tang S, Martirosian V, Ru N, Chmielewski NN, Liang Y, Baulch JE, Acharya MM, Limoli CL. Functional equivalence of stem cell and stem cell-derived extracellular vesicle transplantation to repair the irradiated brain. Stem Cells Transl Med 2020; 9:93-105. [PMID: 31568685 PMCID: PMC6954724 DOI: 10.1002/sctm.18-0227] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 06/17/2019] [Indexed: 01/22/2023] Open
Abstract
Cranial radiotherapy, although beneficial for the treatment of brain tumors, inevitably leads to normal tissue damage that can induce unintended neurocognitive complications that are progressive and debilitating. Ionizing radiation exposure has also been shown to compromise the structural integrity of mature neurons throughout the brain, an effect believed to be at least in part responsible for the deterioration of cognitive health. Past work has shown that cranially transplanted human neural stem cells (hNSCs) or their extracellular vesicles (EVs) afforded long-term beneficial effects on many of these cognitive decrements. To provide additional insight into the potential neuroprotective mechanisms of cell-based regenerative strategies, we have analyzed hippocampal neurons for changes in structural integrity and synaptic remodeling after unilateral and bilateral transplantation of hNSCs or EVs derived from those same cells. Interestingly, hNSCs and EVs similarly afforded protection to host neurons, ameliorating the impact of irradiation on dendritic complexity and spine density for neurons present in both the ipsilateral and contralateral hippocampi 1 month following irradiation and transplantation. These morphometric improvements were accompanied by increased levels of glial cell-derived growth factor and significant attenuation of radiation-induced increases in postsynaptic density protein 95 and activated microglia were found ipsi- and contra-lateral to the transplantation sites of the irradiated hippocampus treated with hNSCs or hNSC-derived EVs. These findings document potent far-reaching neuroprotective effects mediated by grafted stem cells or EVs adjacent and distal to the site of transplantation and support their potential as therapeutic agents to counteract the adverse effects of cranial irradiation.
Collapse
Affiliation(s)
- Sarah M. Smith
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Erich Giedzinski
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Maria C. Angulo
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Tiffany Lui
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Celine Lu
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Audrey L. Park
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Sharon Tang
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Vahan Martirosian
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Ning Ru
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | | | - Yaxuan Liang
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Janet E. Baulch
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Munjal M. Acharya
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| | - Charles L. Limoli
- Department of Radiation OncologyUniversity of CaliforniaIrvineCalifornia
| |
Collapse
|
236
|
Hsu CY, Xiao F, Liu KL, Chen TL, Lee YC, Wang W. Radiomic analysis of magnetic resonance imaging predicts brain metastases velocity and clinical outcome after upfront radiosurgery. Neurooncol Adv 2020; 2:vdaa100. [PMID: 33817641 PMCID: PMC8008166 DOI: 10.1093/noajnl/vdaa100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background Brain metastasis velocity (BMV) predicts outcomes after initial distant brain failure (DBF) following upfront stereotactic radiosurgery (SRS). We developed an integrated model of clinical predictors and pre-SRS MRI-derived radiomic scores (R-scores) to identify high-BMV (BMV-H) patients upon initial identification of brain metastases (BMs). Methods In total, 256 patients with BMs treated with upfront SRS alone were retrospectively included. R-scores were built from 1246 radiomic features in 2 target volumes by using the Extreme Gradient Boosting algorithm to predict BMV-H groups, as defined by BMV at least 4 or leptomeningeal disease at first DBF. Two R-scores and 3 clinical predictors were integrated into a predictive clinico-radiomic (CR) model. Results The related R-scores showed significant differences between BMV-H and low BMV (BMV-L), as defined by BMV less than 4 or no DBF (P < .001). Regression analysis identified BMs number, perilesional edema, and extracranial progression as significant predictors. The CR model using these 5 predictors achieved a bootstrapping corrected C-index of 0.842 and 0.832 in the discovery and test sets, respectively. Overall survival (OS) after first DBF was significantly different between the CR-predicted BMV-L and BMV-H groups (median OS: 26.7 vs 13.0 months, P = .016). Among patients with a diagnosis-specific graded prognostic assessment of 1.5–2 or 2.5–4, the median OS after initial SRS was 33.8 and 67.8 months for CR-predicted BMV-L, compared to 13.5 and 31.0 months for CR-predicted BMV-H (P < .001 and <.001), respectively. Conclusion Our CR model provides a novel approach showing good performance to predict BMV and clinical outcomes.
Collapse
Affiliation(s)
- Che-Yu Hsu
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
- National Taiwan University Cancer Center, Taipei, Taiwan
- Graduate Program of Data Science, National Taiwan University and Academia Sinica, Taipei, Taiwan
- Corresponding Authors: Weichung Wang, PhD, Institute of Applied Mathematical Sciences, National Taiwan University, No.1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan (); Che-Yu Hsu, MD, Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, No. 7, Chung-Shan South Rd, Taipei 100, Taiwan ()
| | - Furen Xiao
- Department of Neurosurgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Kao-Lang Liu
- Department of Medical Imaging, National Taiwan University Cancer Center, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ting-Li Chen
- Graduate Program of Data Science, National Taiwan University and Academia Sinica, Taipei, Taiwan
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
| | - Yueh-Chou Lee
- Department of Mathematics, National Taiwan University, Taipei, Taiwan
| | - Weichung Wang
- Graduate Program of Data Science, National Taiwan University and Academia Sinica, Taipei, Taiwan
- Institute of Applied Mathematical Sciences, National Taiwan University, Taipei, Taiwan
- Corresponding Authors: Weichung Wang, PhD, Institute of Applied Mathematical Sciences, National Taiwan University, No.1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan (); Che-Yu Hsu, MD, Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, No. 7, Chung-Shan South Rd, Taipei 100, Taiwan ()
| |
Collapse
|
237
|
Karunamuni R, Tringale KR, Burkeen J, Tibbs MD, Huynh-Le MP, Bahrami N, Marshall D, Seibert TM, McDonald CR, Hattangadi-Gluth JA. Multi-domain neurocognitive classification of primary brain tumor patients prior to radiotherapy on a prospective clinical trial. J Neurooncol 2020; 146:131-138. [PMID: 31760596 PMCID: PMC7025809 DOI: 10.1007/s11060-019-03353-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/20/2019] [Indexed: 01/10/2023]
Abstract
INTRODUCTION We investigated multi-domain baseline neurocognition of primary brain tumor patients prior to radiotherapy (RT), including clinical predictors of function and association between pre-RT and post-RT impairment on a prospective trial. METHODS A multi-domain neuropsychological battery (memory, executive functioning, language, attention, processing) was performed on 37 patients, pre-RT and 3-(n = 21), 6-(n = 22) and 12-(n = 14) months post-RT. Impairment rate was the proportion of patients with standardized T-scores ≤ 1.5 standard deviations below normative means. Per-patient impairment across all domains was calculated using a global deficit score (GDS; higher value indicates more impairment). Associations between baseline GDS and clinical variables were tested. Global GDS impairment rate at each time point was the fraction of patients with GDS scores > 0.5. RESULTS Statistically significant baseline neurocognitive impairments were identified on 4 memory (all p ≤ 0.03) and 2 out of 3 (p = 0.01, p = 0.027) executive functioning tests. Per-patient baseline GDS was significantly associated with tumor volume (p = 0.048), tumor type (p = 0.043), seizure history (p = 0.007), and use of anti-epileptics (p = 0.009). The percentage of patients with the same impairment status at 3-, 6-, and 12-months as at baseline were 88%, 85%, and 85% respectively. CONCLUSIONS Memory and executive functioning impairment were the most common cognitive deficits prior to RT. Patients with larger tumors, more aggressive histology, and use of anti-epileptics had higher baseline GDS values. GDS is a promising tool to encompass multi-domain neurocognitive function, and baseline GDS can identify those at risk of cognitive impairment.
Collapse
Affiliation(s)
- Roshan Karunamuni
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, 3960 Health Sciences Dr, Mail Code 0865, La Jolla, CA, USA
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA, 92093, USA
| | - Kathryn R Tringale
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, 3960 Health Sciences Dr, Mail Code 0865, La Jolla, CA, USA
| | - Jeffrey Burkeen
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, 3960 Health Sciences Dr, Mail Code 0865, La Jolla, CA, USA
| | - Michelle D Tibbs
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, 3960 Health Sciences Dr, Mail Code 0865, La Jolla, CA, USA
| | - Minh-Phuong Huynh-Le
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, 3960 Health Sciences Dr, Mail Code 0865, La Jolla, CA, USA
| | - Naeim Bahrami
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA, 92093, USA
| | - Deborah Marshall
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, 3960 Health Sciences Dr, Mail Code 0865, La Jolla, CA, USA
| | - Tyler M Seibert
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, 3960 Health Sciences Dr, Mail Code 0865, La Jolla, CA, USA
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA, 92093, USA
| | - Carrie R McDonald
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jona A Hattangadi-Gluth
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, 3960 Health Sciences Dr, Mail Code 0865, La Jolla, CA, USA.
- Center for Multimodal Imaging and Genetics, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
238
|
Turnquist C, Harris BT, Harris CC. Radiation-induced brain injury: current concepts and therapeutic strategies targeting neuroinflammation. Neurooncol Adv 2020; 2:vdaa057. [PMID: 32642709 PMCID: PMC7271559 DOI: 10.1093/noajnl/vdaa057] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Continued improvements in cancer therapies have increased the number of long-term cancer survivors. Radiation therapy remains one of the primary treatment modalities with about 60% of newly diagnosed cancer patients receiving radiation during the course of their disease. While radiation therapy has dramatically improved patient survival in a number of cancer types, the late effects remain a significant factor affecting the quality of life particularly in pediatric patients. Radiation-induced brain injury can result in cognitive dysfunction, including hippocampal-related learning and memory dysfunction that can escalate to dementia. In this article, we review the current understanding of the mechanisms behind radiation-induced brain injury focusing on the role of neuroinflammation and reduced hippocampal neurogenesis. Approaches to prevent or ameliorate treatment-induced side effects are also discussed along with remaining challenges in the field.
Collapse
Affiliation(s)
- Casmir Turnquist
- University of Oxford Medical School, John Radcliffe Hospital, Oxford, UK
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Brent T Harris
- Departments of Neurology and Pathology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
239
|
Karami E, Soliman H, Ruschin M, Sahgal A, Myrehaug S, Tseng CL, Czarnota GJ, Jabehdar-Maralani P, Chugh B, Lau A, Stanisz GJ, Sadeghi-Naini A. Quantitative MRI Biomarkers of Stereotactic Radiotherapy Outcome in Brain Metastasis. Sci Rep 2019; 9:19830. [PMID: 31882597 PMCID: PMC6934477 DOI: 10.1038/s41598-019-56185-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 12/08/2019] [Indexed: 02/08/2023] Open
Abstract
About 20-40% of cancer patients develop brain metastases, causing significant morbidity and mortality. Stereotactic radiation treatment is an established option that delivers high dose radiation to the target while sparing the surrounding normal tissue. However, up to 20% of metastatic brain tumours progress despite stereotactic treatment, and it can take months before it is evident on follow-up imaging. An early predictor of radiation therapy outcome in terms of tumour local failure (LF) is crucial, and can facilitate treatment adjustments or allow for early salvage treatment. In this study, an MR-based radiomics framework was proposed to derive and investigate quantitative MRI (qMRI) biomarkers for the outcome of LF in brain metastasis patients treated with hypo-fractionated stereotactic radiation therapy (SRT). The qMRI biomarkers were constructed through a multi-step feature extraction/reduction/selection framework using the conventional MR imaging data acquired from 100 patients (133 lesions), and were applied in conjunction with machine learning techniques for outcome prediction and risk assessment. The results indicated that the majority of the features in the optimal qMRI biomarkers characterize the heterogeneity in the surrounding regions of tumour including edema and tumour/lesion margins. The optimal qMRI biomarker consisted of five features that predict the outcome of LF with an area under the curve (AUC) of 0.79, and a cross-validated sensitivity and specificity of 81% and 79%, respectively. The Kaplan-Meier analyses showed a statistically significant difference in local control (p-value < 0.0001) and overall survival (p = 0.01). Findings from this study are a step towards using qMRI for early prediction of local failure in brain metastasis patients treated with SRT. This may facilitate early adjustments in treatment, such as surgical resection or salvage radiation, that can potentially improve treatment outcomes. Investigations on larger cohorts of patients are, however, required for further validation of the technique.
Collapse
Affiliation(s)
- Elham Karami
- Department of Electrical Engineering and Computer Science, Lassonde School of Engineering, York University, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Hany Soliman
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Mark Ruschin
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Arjun Sahgal
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Sten Myrehaug
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Chia-Lin Tseng
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Gregory J Czarnota
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | | | - Brige Chugh
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
| | - Angus Lau
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Greg J Stanisz
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University, Lublin, Poland
| | - Ali Sadeghi-Naini
- Department of Electrical Engineering and Computer Science, Lassonde School of Engineering, York University, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
- Physical Sciences Platform, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.
| |
Collapse
|
240
|
Tringale KR, Nguyen TT, Karunamuni R, Seibert T, Huynh-Le MP, Connor M, Moiseenko V, Gorman MK, Marshall A, Tibbs MD, Farid N, Simpson D, Sanghvi P, McDonald CR, Hattangadi-Gluth JA. Quantitative Imaging Biomarkers of Damage to Critical Memory Regions Are Associated With Post-Radiation Therapy Memory Performance in Brain Tumor Patients. Int J Radiat Oncol Biol Phys 2019; 105:773-783. [PMID: 31408667 PMCID: PMC6876859 DOI: 10.1016/j.ijrobp.2019.08.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 05/23/2019] [Accepted: 08/05/2019] [Indexed: 01/06/2023]
Abstract
PURPOSE We used quantitative magnetic resonance imaging to prospectively analyze the association between microstructural damage to memory-associated structures within the medial temporal lobe and longitudinal memory performance after brain radiation therapy (RT). METHODS AND MATERIALS Patients with a primary brain tumor receiving fractionated brain RT were enrolled on a prospective trial (n = 27). Patients underwent high-resolution volumetric brain magnetic resonance imaging, diffusion-weighted imaging, and neurocognitive testing before and 3, 6, and 12 months post-RT. Medial temporal lobe regions (hippocampus; entorhinal, parahippocampal, and temporal pole white matter [WM]) were autosegmented, quantifying volume and diffusion biomarkers of WM integrity (mean diffusivity [MD]; fractional anisotropy [FA]). Reliable change indices measured changes in verbal (Hopkins Verbal Learning Test-Revised) and visuospatial (Brief Visuospatial Memory Test-Revised [BVMT-R]) memory. Linear mixed-effects models assessed longitudinal associations between imaging parameters and memory. RESULTS Visuospatial memory significantly declined at 6 months post-RT (mean reliable change indices, -1.3; P = .012). Concurrent chemotherapy and seizures trended toward a significant association with greater decline in visuospatial memory (P = .053 and P = .054, respectively). Higher mean dose to the left temporal pole WM was significantly associated with decreased FA (r = -0.667; P = .002). Over all time points, smaller right hippocampal volume (P = .021), lower right entorhinal FA (P = .023), greater right entorhinal MD (P = .047), and greater temporal pole MD (BVMT-R total recall, P = .003; BVMT-R delayed recall, P = .042) were associated with worse visuospatial memory. The interaction between right entorhinal MD (BVMT-R total recall, P = .021; BVMT-R delayed recall, P = .004) and temporal pole FA (BVMT-R delayed recall, P = .024) significantly predicted visuospatial memory performance. CONCLUSIONS Brain tumor patients exhibited visuospatial memory decline post-RT. Microstructural damage to critical memory regions, including the hippocampus and medial temporal lobe WM, were associated with post-RT memory decline. The integrity of medial temporal lobe structures is critical to memory performance post-RT, representing possible avoidance targets for memory preservation.
Collapse
Affiliation(s)
- Kathryn R Tringale
- Department of Radiation Medicine and Applied Sciences; Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Roshan Karunamuni
- Department of Radiation Medicine and Applied Sciences; Center for Multimodal Imaging and Genetics
| | - Tyler Seibert
- Department of Radiation Medicine and Applied Sciences; Center for Multimodal Imaging and Genetics
| | | | | | | | | | | | | | - Nikdokht Farid
- Department of Radiology, University of California, San Diego, La Jolla, California
| | | | - Parag Sanghvi
- Department of Radiation Medicine and Applied Sciences
| | - Carrie R McDonald
- Department of Radiation Medicine and Applied Sciences; Department of Psychiatry; Center for Multimodal Imaging and Genetics
| | - Jona A Hattangadi-Gluth
- Department of Radiation Medicine and Applied Sciences; Center for Multimodal Imaging and Genetics.
| |
Collapse
|
241
|
Fecci PE, Champion CD, Hoj J, McKernan CM, Goodwin CR, Kirkpatrick JP, Anders CK, Pendergast AM, Sampson JH. The Evolving Modern Management of Brain Metastasis. Clin Cancer Res 2019; 25:6570-6580. [PMID: 31213459 PMCID: PMC8258430 DOI: 10.1158/1078-0432.ccr-18-1624] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/23/2019] [Accepted: 06/13/2019] [Indexed: 12/11/2022]
Abstract
The incidence of brain metastases is increasing as cancer therapies improve and patients live longer, providing new challenges to the multidisciplinary teams that care for these patients. Brain metastatic cancer cells possess unique characteristics that allow them to penetrate the blood-brain barrier, colonize the brain parenchyma, and persist in the intracranial environment. In addition, brain metastases subvert the innate and adaptive immune system, permitting evasion of the antitumor immune response. Better understanding of the above mechanisms will allow for development and delivery of more effective therapies for brain metastases. In this review, we outline the molecular mechanisms underlying development, survival, and immunosuppression of brain metastases. We also discuss current and emerging treatment strategies, including surgery, radiation, disease-specific and mutation-targeted systemic therapy, and immunotherapy.
Collapse
Affiliation(s)
- Peter E Fecci
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- Duke Center for Brain and Spinal Metastases, Duke University Medical Center, Durham, North Carolina
| | - Cosette D Champion
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- Duke Center for Brain and Spinal Metastases, Duke University Medical Center, Durham, North Carolina
| | - Jacob Hoj
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Courtney M McKernan
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - C Rory Goodwin
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- Duke Center for Brain and Spinal Metastases, Duke University Medical Center, Durham, North Carolina
| | - John P Kirkpatrick
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina
- Duke Center for Brain and Spinal Metastases, Duke University Medical Center, Durham, North Carolina
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - Carey K Anders
- Duke Cancer Institute, Division of Medical Oncology, Duke University Medical Center, Durham, North Carolina
| | - Ann Marie Pendergast
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - John H Sampson
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina.
- Duke Center for Brain and Spinal Metastases, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
242
|
Radiation chronotherapy-clinical impact of treatment time-of-day: a systematic review. J Neurooncol 2019; 145:415-427. [PMID: 31729636 DOI: 10.1007/s11060-019-03332-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 11/03/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE Many brain tumor patients suffer from radiation-induced toxicities. Chronotherapy is a treatment modality that utilizes circadian rhythms to optimize the effect on tumor while minimizing negative outcomes on healthy tissue. This review aims to systematically examine the literature on the application of a radiation chronotherapeutic for all cancers and determine the possible advantages of incorporating a circadian-based fixed time-of-day for radiotherapy into CNS cancers. METHODS A systematic review of the literature was conducted in two electronic databases from inception to February 1, 2019. Primary research manuscripts were screened for those related to adult human subjects exposed to ionizing radiation using the chronotherapy technique. RESULTS Nine manuscripts were included in the review from 79 eligible articles. Three were prospective randomized trails and 6 were retrospective reviews. This survey revealed that overall survival and tumor control do not have consistent effects with only 60% and 55.5% of paper which included the variables having some significance, respectively. Treatment symptoms were the primary endpoint for both the prospective trials and were examined in 3 of the retrospective reviews; effects were observed in sensitive tissue for all 5 studies including mucosal linings and skin basal layer. CONCLUSIONS Existing literature suggests that the application of radiation chronotherapy may reduce negative symptom outcome within highly proliferative tissues. Further examination of radiation chronotherapy in well-designed prospective trials and studies in brain tumor patients are merited.
Collapse
|
243
|
Kang KJ, Jung KH, Choi EJ, Kim H, Do SH, Ko IO, Oh SJ, Lee YJ, Kim JY, Park JA. Monitoring Physiological Changes in Neutron-Exposed Normal Mouse Brain Using FDG-PET and DW-MRI. Radiat Res 2019; 193:54-62. [PMID: 31682543 DOI: 10.1667/rr15405.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
We monitored a physiological response in a neutron-exposed normal mouse brain using two imaging tools, [18F]fluro-deoxy-D-glucose positron emission tomography ([18F]FDG-PET) and diffusion weighted-magnetic resonance imaging (DW-MRI), as an imaging biomarker. We measured the apparent diffusion coefficient (ADC) of DW-MRI and standardized uptake value (SUV) of [18F]FDG-PET, which indicated changes in the cellular environment for neutron irradiation. This approach was sensitive enough to detect cell changes that were not confirmed in hematoxylin and eosin (H&E) results. Glucose transporters (GLUT) 1 and 3, indicators of the GLUT capacity of the brain, were significantly decreased after neutron irradiation, demonstrating that the change in blood-brain-barrier (BBB) permeability affects the GLUT, with changes in both SUV and ADC values. These results demonstrate that combined imaging of the same object can be used as a quantitative indicator for in vivo pathological changes. In particular, the radiation exposure assessment of combined imaging, with specific integrated functions of [18F]FDG-PET and MRI, can be employed repeatedly for noninvasive analysis performed in clinical practice. Additionally, this study demonstrated a novel approach to assess the extent of damage to normal tissues as well as therapeutic effects on tumors.
Collapse
Affiliation(s)
- Kyung Jun Kang
- Division of Applied RI, Korea Institute Radiological and Medical Sciences, Seoul, Korea 01812
| | - Ki-Hye Jung
- Division of Applied RI, Korea Institute Radiological and Medical Sciences, Seoul, Korea 01812
| | - Eun-Ji Choi
- College of Veterinary Medicine, Konkuk University, Seoul, Korea 05029
| | - Hyosung Kim
- College of Veterinary Medicine, Konkuk University, Seoul, Korea 05029
| | - Sun Hee Do
- College of Veterinary Medicine, Konkuk University, Seoul, Korea 05029
| | - In Ok Ko
- Division of Applied RI, Korea Institute Radiological and Medical Sciences, Seoul, Korea 01812
| | - Se Jong Oh
- Division of Applied RI, Korea Institute Radiological and Medical Sciences, Seoul, Korea 01812
| | - Yong Jin Lee
- Division of Applied RI, Korea Institute Radiological and Medical Sciences, Seoul, Korea 01812
| | - Jung Young Kim
- Division of Applied RI, Korea Institute Radiological and Medical Sciences, Seoul, Korea 01812
| | - Ji-Ae Park
- Division of Applied RI, Korea Institute Radiological and Medical Sciences, Seoul, Korea 01812
| |
Collapse
|
244
|
Jost G, Frenzel T, Boyken J, Pietsch H. Impact of brain tumors and radiotherapy on the presence of gadolinium in the brain after repeated administration of gadolinium-based contrast agents: an experimental study in rats. Neuroradiology 2019; 61:1273-1280. [PMID: 31297571 PMCID: PMC6817760 DOI: 10.1007/s00234-019-02256-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 07/01/2019] [Indexed: 02/04/2023]
Abstract
PURPOSE To investigate the impact of blood-brain barrier (BBB) alterations induced by an experimental tumor and radiotherapy on MRI signal intensity (SI) in deep cerebellar nuclei (DCN) and the presence of gadolinium after repeated administration of a linear gadolinium-based contrast agent in rats. METHODS Eighteen Fischer rats were divided into a tumor (gliosarcoma, GS9L model), a radiotherapy, and a control group. All animals received 5 daily injections (1.8 mmol/kg) of gadopentetate dimeglumine. For tumor-bearing animals, the BBB disruption was confirmed by contrast-enhanced MRI. Animals from the tumor and radiation group underwent radiotherapy in 6 fractions of 5 Gray. The SI ratio between DCN and brain stem was evaluated on T1-weigthed MRI at baseline and 1 week after the last administration. Subsequently, the brain was dissected for gadolinium quantification by inductively coupled plasma-mass spectrometry. Statistical analysis was done with the Kruskal-Wallis test. RESULTS An increased but similar DCN/brain stem SI ratio was found for all three groups (p = 0.14). The gadolinium tissue concentrations (median, nmol/g) were 6.7 (tumor), 6.3 (radiotherapy), and 6.8 (control) in the cerebellum (p = 0.64) and 17.8/14.6 (tumor), 20.0/18.9 (radiotherapy), and 17.8/15.9 (control) for the primary tumor (p = 0.98) and the contralateral hemisphere (p = 0.41) of the cerebrum, respectively. CONCLUSION An experimental brain tumor treated by radiotherapy or radiotherapy alone did not alter DCN signal hyperintensity and gadolinium concentration in the rat brain 1 week after repeated administration of gadopentetate. This suggests that a local BBB disruption does not affect the amount of retained gadolinium in the brain.
Collapse
Affiliation(s)
- Gregor Jost
- Bayer AG, MR & CT Contrast Media Research, Muellerstrasse 178, 13353, Berlin, Germany.
| | - Thomas Frenzel
- Bayer AG, MR & CT Contrast Media Research, Muellerstrasse 178, 13353, Berlin, Germany
| | - Janina Boyken
- Bayer AG, MR & CT Contrast Media Research, Muellerstrasse 178, 13353, Berlin, Germany
| | - Hubertus Pietsch
- Bayer AG, MR & CT Contrast Media Research, Muellerstrasse 178, 13353, Berlin, Germany
| |
Collapse
|
245
|
Abstract
INTRODUCTION Hyperbaric oxygen therapy (HBOT) shows promising results in treating radionecrosis (RN) but there is limited evidence for its use in brain RN. The purpose of this study is to report the outcomes of using HBOT for symptomatic brain RN at a single institution. METHODS This was a retrospective review of patients with symptomatic brain RN between 2008 and 2018 and was treated with HBOT. Demographic data, steroid use, clinical response, radiologic response and toxicities were collected. The index time for analysis was the first day of HBOT. The primary endpoint was clinical improvement of a presenting symptom, including steroid dose reduction. RESULTS Thirteen patients who received HBOT for symptomatic RN were included. The median time from last brain radiation therapy to presenting symptoms of brain RN was 6 months. Twelve patients (92%) had clinical improvement with median time to symptom improvement of 33 days (range 1-109 days). One patient had transient improvement after HBOT but had recurrent symptomatic RN at 12 months. Of the eight patients with evaluable follow-up MRI, four patients had radiological improvement while four had stable necrosis appearance. Two patients had subsequent deterioration in MRI appearances, one each in the background of initial radiologic improvement and stability. Median survival was 15 months with median follow-up of 10 months. Seven patients reported side effects attributable to HBOT (54%), four of which were otologic in origin. CONCLUSIONS HBOT is a safe and effective treatment for brain RN. HBOT showed clinical and radiologic improvement or stability in most patients. Prospective studies to further evaluate the effectiveness and side effects of HBOT are needed.
Collapse
|
246
|
Zhang D, Zhou W, Lam TT, Weng C, Bronk L, Ma D, Wang Q, Duman JG, Dougherty PM, Grosshans DR. Radiation induces age-dependent deficits in cortical synaptic plasticity. Neuro Oncol 2019; 20:1207-1214. [PMID: 29660023 DOI: 10.1093/neuonc/noy052] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Radiation-induced cognitive dysfunction is a significant side effect of cranial irradiation for brain tumors. Clinically, pediatric patients are more vulnerable than adults. However, the underlying mechanisms of dysfunction, including reasons for age dependence, are still largely unknown. Previous studies have focused on the loss of hippocampal neuronal precursor cells and deficits in memory. However, survivors may also experience deficits in attention, executive function, or other non-hippocampal-dependent cognitive domains. We hypothesized that brain irradiation induces age-dependent deficits in cortical synaptic plasticity. Methods In vivo recordings were used to test neuronal plasticity along the direct pathway from the cornu ammonis 1 (CA1)/subicular region to the prefrontal cortex (PFC). Specifically, long-term potentiation (LTP) in the CA1/subicular-PFC pathway was assessed after cranial irradiation of juvenile and adult Sprague Dawley rats. We further assessed a potential role for glutamate toxicity by evaluating the potential neuroprotective effects of memantine. Results LTP was greatly inhibited in both adult and juvenile animals at 3 days after radiation but returned to near-normal levels by 8 weeks-only in adult rats. Memantine given before, but not after, irradiation partially prevented LTP inhibition in juvenile and adult rats. Conclusion Cranial radiation impairs neuroplasticity along the hippocampal-PFC pathway; however, its effects vary by age. Pretreatment with memantine offered protection to both juvenile and adult animals. Deficits in cortical plasticity may contribute to radiation-induced cognitive dysfunction, including deficits in attention and age-dependent sensitivity of such pathways, which may underlie differences in clinical outcomes between juveniles and adults after cranial irradiation.
Collapse
Affiliation(s)
- Die Zhang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wei Zhou
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Thanh Thai Lam
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Connie Weng
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lawrence Bronk
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Duo Ma
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Qiang Wang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joseph G Duman
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas
| | - Patrick M Dougherty
- Departments of Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David R Grosshans
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
247
|
Di Giacomo AM, Valente M, Cerase A, Lofiego MF, Piazzini F, Calabrò L, Gambale E, Covre A, Maio M. Immunotherapy of brain metastases: breaking a "dogma". J Exp Clin Cancer Res 2019; 38:419. [PMID: 31623643 PMCID: PMC6798349 DOI: 10.1186/s13046-019-1426-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 09/23/2019] [Indexed: 12/18/2022] Open
Abstract
Until very few years ago, the oncology community dogmatically excluded any clinical potential for immunotherapy in controlling brain metastases. Therefore, despite the significant therapeutic efficacy of monoclonal antibodies to immune check-point(s) across a wide range of tumor types, patients with brain disease were invariably excluded from clinical trials with these agents. Recent insights on the immune landscape of the central nervous system, as well as of the brain tumor microenvironment, are shedding light on the immune-biology of brain metastases.Interestingly, retrospective analyses, case series, and initial prospective clinical trials have recently investigated the role of different immune check-point inhibitors in brain metastases, reporting a significant clinical activity also in this subset of patients. These findings, and their swift translation in the daily practice, are driving fundamental changes in the clinical management of patients with brain metastases, and raise important neuroradiologic challenges. Along this line, neuro-oncology undoubtedly represents an additional area of active investigation and of growing interest to support medical oncologists in the evaluation of clinical responses of brain metastases to ICI treatment, and in the management of neurologic immune-related adverse events.Aim of this review is to summarize the most recent findings on brain metastases immunobiology, on the evolving scenario of clinical efficacy of ICI therapy in patients with brain metastases, as well as on the increasing relevance of neuroradiology in this therapeutic setting.
Collapse
Affiliation(s)
- Anna Maria Di Giacomo
- Department of Oncology, Center for Immuno-Oncology, Medical Oncology and Immunotherapy, University Hospital of Siena, Viale Bracci, 14, 53100 Siena, Italy
| | - Monica Valente
- Department of Oncology, Center for Immuno-Oncology, Medical Oncology and Immunotherapy, University Hospital of Siena, Viale Bracci, 14, 53100 Siena, Italy
| | - Alfonso Cerase
- Unit of Neuroradiology, University Hospital, Siena, Italy
| | - Maria Fortunata Lofiego
- Department of Oncology, Center for Immuno-Oncology, Medical Oncology and Immunotherapy, University Hospital of Siena, Viale Bracci, 14, 53100 Siena, Italy
| | - Francesca Piazzini
- Department of Oncology, Center for Immuno-Oncology, Medical Oncology and Immunotherapy, University Hospital of Siena, Viale Bracci, 14, 53100 Siena, Italy
| | - Luana Calabrò
- Department of Oncology, Center for Immuno-Oncology, Medical Oncology and Immunotherapy, University Hospital of Siena, Viale Bracci, 14, 53100 Siena, Italy
| | - Elisabetta Gambale
- Department of Oncology, Center for Immuno-Oncology, Medical Oncology and Immunotherapy, University Hospital of Siena, Viale Bracci, 14, 53100 Siena, Italy
| | - Alessia Covre
- Department of Oncology, Center for Immuno-Oncology, Medical Oncology and Immunotherapy, University Hospital of Siena, Viale Bracci, 14, 53100 Siena, Italy
| | - Michele Maio
- Department of Oncology, Center for Immuno-Oncology, Medical Oncology and Immunotherapy, University Hospital of Siena, Viale Bracci, 14, 53100 Siena, Italy
| |
Collapse
|
248
|
Carnell LS, Homer M, Hoots K, Meeks H, Prasanna PGS, Rios C, Simonsen LC, Taliaferro LP, Wathen LK. Medical countermeasures for radiation induced health effects: report of an Interagency Panel Session held at the NASA Human Research Program Investigator's Workshop, 26 January 2017. Int J Radiat Biol 2019; 97:S117-S124. [PMID: 31490103 PMCID: PMC8463010 DOI: 10.1080/09553002.2019.1665214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
An Interagency Panel Session organized by the NASA Human Research Program (HRP) Space Radiation Program Element (SRPE) was held during the NASA HRP Investigator’s Workshop (IWS) in Galveston, Texas on 26 January 2017 to identify complementary research areas that will advance the testing and development of medical countermeasures (MCMs) in support of radioprotection and radiation mitigation on the ground and in space. There were several areas of common interest identified among the various participating agencies. This report provides a summary of the topics discussed by each agency along with potential areas of intersection for mutual collaboration opportunities. Common goals induded repurposing of pharmaceuticals, nutraceuticals for use as radioprotectors and/or mitigators, low-dose/chronic exposure paradigms, late effects post-radiation exposure, mixed-field exposures of gamma-neutron, performance decrements, and methods to determine individual exposure levels.
Collapse
Affiliation(s)
| | - Mary Homer
- US Department of Health and Human Services, Biomedical Advanced Research and Development Authority, Office of the Assistant Secretary for Preparedness and Response, Washington, DC, USA
| | - Keith Hoots
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Heather Meeks
- Defense Threat Reduction Agency, Fort Belvoir, VA, USA
| | - Pataje G S Prasanna
- US Department of Health and Human Services, Division of Cancer Treatment and Diagnosis, Radiation Research Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Carmen Rios
- Division of Allergy, Immunology and Transplantation (DAIT), Radiation and Nuclear Countermeasures Program (RNCP), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD, USA
| | | | - Lanyn P Taliaferro
- Division of Allergy, Immunology and Transplantation (DAIT), Radiation and Nuclear Countermeasures Program (RNCP), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD, USA
| | - Lynne K Wathen
- US Department of Health and Human Services, Biomedical Advanced Research and Development Authority, Office of the Assistant Secretary for Preparedness and Response, Washington, DC, USA
| |
Collapse
|
249
|
McTyre ER, Soike MH, Farris M, Ayala-Peacock DN, Hepel JT, Page BR, Shen C, Kleinberg L, Contessa JN, Corso C, Chiang V, Henson-Masters A, Cramer CK, Ruiz J, Pasche B, Watabe K, D'Agostino R, Su J, Laxton AW, Tatter SB, Fiveash JB, Ahluwalia M, Kotecha R, Chao ST, Braunstein SE, Attia A, Chung C, Chan MD. Multi-institutional validation of brain metastasis velocity, a recently defined predictor of outcomes following stereotactic radiosurgery. Radiother Oncol 2019; 142:168-174. [PMID: 31526671 DOI: 10.1016/j.radonc.2019.08.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Brain metastasis velocity (BMV) is a prognostic metric that describes the recurrence rate of new brain metastases after initial treatment with radiosurgery (SRS). We have previously risk stratified patients into high, intermediate, and low-risk BMV groups, which correlates with overall survival (OS). We sought to externally validate BMV in a multi-institutional setting. METHODS Patients from nine academic centers were treated with upfront SRS; the validation cohort consisted of data from eight institutions not previously used to define BMV. Patients were classified by BMV into low (<4 BMV), intermediate (4-13 BMV), and high-risk groups (>13 BMV). Time-to-event outcomes were estimated using the Kaplan-Meier method. Cox proportional hazards methods were used to estimate the effect of BMV and salvage modality on OS. RESULTS Of 2829 patients, 2092 patients were included in the validation dataset. Of these, 921 (44.0%) experienced distant brain failure (DBF). Median OS from initial SRS was 11.2 mo. Median OS for BMV < 4, BMV 4-13, and BMV > 13 were 12.5 mo, 7.0 mo, and 4.6 mo (p < 0.0001). After multivariate regression modeling, melanoma histology (β: 10.10, SE: 1.89, p < 0.0001) and number of initial brain metastases (β: 1.52, SE: 0.34, p < 0.0001) remained predictive of BMV (adjusted R2 = 0.06). CONCLUSIONS This multi-institutional dataset validates BMV as a predictor of OS following initial SRS. BMV is being utilized in upcoming multi-institutional randomized controlled trials as a stratification variable for salvage whole brain radiation versus salvage SRS after DBF.
Collapse
Affiliation(s)
- Emory R McTyre
- Department of Radiation Oncology, Greenville Health System Cancer Institute, USA
| | - Michael H Soike
- Hazelrig-Salter Radiation Oncology Center, University of Alabama at Birmingham, USA.
| | - Michael Farris
- Department of Radiation Oncology, Wake Forest School of Medicine, USA
| | | | - Jaroslaw T Hepel
- Department of Radiation Oncology, Brown University Alpert Medical School, USA
| | - Brandi R Page
- Department of Radiation Oncology, Johns Hopkins School of Medicine, USA
| | - Colette Shen
- Department of Radiation Oncology, University of North Carolina, USA
| | | | | | | | | | | | | | - Jimmy Ruiz
- Department of Medicine, Wake Forest School of Medicine, USA
| | - Boris Pasche
- Department of Medicine, Wake Forest School of Medicine, USA
| | - Kounosuke Watabe
- Department of Cancer Biology, Wake Forest School of Medicine, USA
| | - Ralph D'Agostino
- Department of Biostatistical Sciences, Wake Forest School of Medicine, USA
| | - Jing Su
- Department of Biostatistical Sciences, Wake Forest School of Medicine, USA
| | - Adrian W Laxton
- Department of Neurosurgery, Wake Forest School of Medicine, USA
| | | | - John B Fiveash
- Hazelrig-Salter Radiation Oncology Center, University of Alabama at Birmingham, USA
| | - Manmeet Ahluwalia
- Brain Tumor and Neuro-Oncology Center, Neurological Institute, Cleveland Clinic Foundation, USA
| | - Rupesh Kotecha
- Department of Radiation Oncology, Miami Cancer Institute, USA
| | - Samuel T Chao
- Brain Tumor and Neuro-Oncology Center, Neurological Institute, Cleveland Clinic Foundation, USA
| | - Steve E Braunstein
- Department of Radiation Oncology, University of California San Francisco, USA
| | - Albert Attia
- Department of Radiation Oncology, Vanderbilt University School of Medicine, USA
| | - Caroline Chung
- Department of Radiation Oncology, M.D. Anderson Cancer Center, USA
| | - Michael D Chan
- Department of Radiation Oncology, Wake Forest School of Medicine, USA
| |
Collapse
|
250
|
Medulloblastoma in childhood: What effects on neurocognitive functions? Cancer Radiother 2019; 23:370-377. [DOI: 10.1016/j.canrad.2018.11.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 11/13/2018] [Accepted: 11/20/2018] [Indexed: 11/23/2022]
|