Skip to main content
Dose-Response logoLink to Dose-Response
. 2020 Jul 9;18(3):1559325820938279. doi: 10.1177/1559325820938279

Biomarkers of Brain Damage Induced by Radiotherapy

Nahida Sultana 1,, Chao Sun 2, Takanori Katsube 3, Bing Wang 3,
PMCID: PMC7350401  PMID: 32694960

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.

Keywords: biomarker, cranial radiotherapy, radiation-induced brain damage, cognitive impairment

Introduction

Brain tumors are one of the leading causes of cancer-related death especially in children.1-3 The metastatic brain tumors, generally from lung carcinoma, breast carcinoma, and melanoma, experienced by about 10% to 30% of adult patients with cancer and 6% to 10% of children with cancer, are the main reason of morbidity and mortality.4 Radiotherapy (RT) remains currently a critical component for both primary and metastatic brain tumors either alone or in combination with surgery, chemotherapy, and molecularly targeted agents. However, simultaneously it causes normal brain tissue injury which leads to serious health consequences. Previously it was considered that the brain was the major radioresistant part of the body but now it has been proved and accepted that the brain is one of the most radiosensitive organs in the clinical RT.5 Recent studies reported that cranial RT is the major cause of cognitive impairments and other complications of the brain.6,7 As a fact, radiation-induced detrimental effects on normal brain tissue limit the benefit of RT for the treatment of brain tumors.8-13

Due to the late health consequences of cranial RT, treatment of brain tumors has become more complicated in many aspects. For example, predicting individual radiosensitivity, which can differ from hypersensitivity to resistance depending on both individual genotype and tumor type, exact delivery of the radiation dose, realization of the exposure mode, and clinical limitation for diagnosis of radiation-induced necrosis from continued tumor growth.14 Thus, to improve treatment outcome and the quality of life of the patient, understanding of the damage and the underlying mechanisms is essential for identifying potential opportunities to protect the patient from severe damage and/or mitigate the detrimental effects. In this context, biomarker studies introduce a novel era for early diagnosis and ensuring effective treatment. A biomarker is a characteristic that is objectively measured and evaluated as an indicator of normal biological processes, pathogenic processes, or pharmacologic responses to a therapeutic intervention.15 A biomarker can accurately indicate the actual biological, pathological, and therapeutic condition of the host. A predictive biomarker specifies the benefit or the outcome to the patient from the treatment, assessed to their condition at baseline.16 Studies of biomarkers, in particular predictive biomarkers, for radiation-induced brain injury and translation of the research advances to RT would enable stratifying patients for customized treatment and improving therapeutic efficacy and the quality of life.

In this minireview, we would give a brief overview on the current main cranial RT based on the latest literatures, analyze the documented work on the acute brain damage and late consequences induced by RT, try to identify the biomarkers, in particular, the predictive biomarkers for the damage, and summarize the biological and clinical significance of the biomarkers.

Cranial RT

Both primary and metastatic brain tumors are of the most combative and damaging forms of cancer. Although the exact etiology is still unknown, various genetic and environmental risk factors were identified.17 Treatment actions for brain tumors are mainly depend on the type, location, size, and grade of the tumor, and age and health conditions of the patient. Generally, surgery, chemotherapy, and RT are accepted as standard treatment procedures for brain tumors. Being effective for accessible and single area of tumor, surgery is usually the initial treatment step for most primary and malignant brain tumors, while it is ineffectual for all types of malignant tumors. Chemotherapy acts as an adjuvant with the combination of surgery and RT but an effective treatment procedure for brain tumors due to the drug restriction by the blood–brain barrier (BBB). Radiotherapy, as one of the standard treatment option for brain tumors, applying controlled high energetic ionizing radiation (IR) such as X-ray and γ-ray could either damage cancer cells directly or arrest cell cycle to limit their ability to grow.18 Ionizing radiation can be administered externally and internally, and external RT is an important way to treat brain tumors in many patients.19

Whole-Brain Radiation Therapy

Whole-brain radiation therapy (WBRT) is externally delivered to the entire brain, which is considered a well-advised treatment option for multiple brain metastases. It is more effective than surgery and stereotactic radiosurgery (SRS), destroying both gross and microscopic tumors. It is also used simultaneously with surgery and SRS to ameliorate local control.20 In addition, WBRT is applicable to treat patients with metastases invading important the part of the brain or patients ineffective from either surgery or SRS. Currently, the most practice radiation dose for brain metastases is 30 Gy in 10 fractions over 2 weeks. It was reported that WBRT could achieve an average survival of 3 to 6 months,21-24 decrease the recurrence incidence of metastases, reduce death from neurological damages,25 and improve the quality of life in 75% to 85% of patients by controlling and upgrading neurological symptoms.23 On the other hand, some studies showed that WBRT could retard tumor growth but eliminating the tumor, neither increasing the period of functional independence and overall survival rate.26 Cognitive deficits are considered the side effects of WBRT that adversely affects the quality of life. As a fact, cognitive impairments were observed in 50% to 90% of adult patients with brain tumor 6 months after WBRT.27-29 In addition, delayed disintegration of cognitive function,30 growth hormone deficiency, and motor dysfunction were also observed in laboratory and clinical studies.31

Stereotactic Radiosurgery

As an alternative option to neurosurgery, in SRS, high energy X-rays, γ-rays, or protons are delivered in a single large dose or a few large doses to a surgically inaccessible discrete tumor. Multiple convergent beams are used to attenuate high dose exposure of normal tissue. Stereotactic radiosurgery is applied to treat a single tumor or multiple tumors (usually up to 3) and can be effectively used to treat deep intercranial surgically inaccessible lesions. Retroactive studies showed that SRS seemed to be equivalent to surgery32,33 and highly potential in inhibiting progressive tumor growth.34 From this aspect, studies exhibited survival value enhancement as well as progress in Karnofsky Performance Status using SRS after WBRT.32,35 The limitation of this noninvasive treatment technique is that it was only advised or suitable for small tumors less than 3 cm in diameter and radiographically well-defined tumors by computed tomography or magnetic resonance scans. Furthermore, the expected dosage of radiation may not be safely delivered to the cancerous brain tumor due to the close proximity to the sensitive and critical normal portion of the brain, such as the optic nerve, hippocampus, and spinal cord or bowel.

Radiation-Induced Acute Brain Damage and Late Health Consequences

Cranial RT is extensively used to treat tumor growth and propagation, and IR could simultaneously affect the normal tissue of the central nervous system (CNS) via direct action (hitting the biomolecule of the cell, disrupting the molecular structure, particularly DNA) and indirect action (producing highly reactive free radical atoms and interacting free radicals with brain molecules), causing damage or the structural alteration or even cell death.36 Compared to early responding tissues such as bone marrow, the vascular tissues, nerves, and parenchyma in the brain, and spinal cord are late responding to radiation exposure and usually do not manifest instantly radiation-induced effects, for example, vascular abnormalities, demyelination, irreversible necrosis of the white matter (WM), permeability changes in the BBB, reduced amount of endothelial cells, injury of oligodendrocyte precursor cells, and activation of astrocytes and microglia,14,37 leading to delayed neurological difficulties and neurocognitive shortages in long-term survivors.38 The late neurological dysfunction includes functional and cognitive impairments, with deficits in learning, attention, working memory, verbal memory, executive function, vision, motor function, severe dementia, and eventually the quality of life of the patients. The occurrence of radiation-induced brain damage after conventional RT was 5% to 24%.39 Of note, the cognitive impairments could occur without the appearance of any structural modifications in the brain tissue.40

Radiation-induced brain injury could be classified into 3 phases: acute, early delayed, and late-delayed injury.38 Acute brain injury occurs in days to weeks after WBRT and SRS, involving fatigue, hair loss, skin erythema, headache, nausea, and lethargy, which is unusual with the present RT techniques. Early delayed brain injury is observed within 1 to 6 months post RT, showing fatigue, somnolence, short-term memory loss, and temporary demyelination. Clinically late-delayed injury is associated with the symptoms of vascular abnormalities, demyelination, and eventually WM necrosis,41 commonly observed from 6 months to several years after RT, and these late-delayed damages are considered as irreversible and progressive. In most of the cases, the cognitive deficiency was the consequence of radiation-induced late delayed injury42 presently occurring in 50% to 90% of brain tumor survivors43,44 and amplifying with the improvement of RT techniques.45,46 The advancing degeneration affected the physical and mental health and declined the quality of life of the long-term survivors.47,48 It is well-documented that high radiation doses (>60 Gy) were responsible for permanent injury, while emerging analysis also exhibited that low radiation doses (<20 Gy) could also arise late delayed damage.49 Of particular concern, both acute and early delayed symptoms and damages were typically reversible that could be solved automatically but late delayed injury, which was counted as permanent brain damage.47,48,50 Currently, our knowledge on the mechanisms underlying radiation-induced cellular and molecular brain injury was still limited, while cumulated data indicated that radiation-induced damage in cerebral tissues was an extremely complex and interactive way associated with various components of tissues.5,38,51 Studies showed that radiation-induced memory losses and attention deficits were associated with neuroinflammation, BBB alterations, and demyelination, in addition to decreased neurogenesis.52-60 Cerebral vascular tissue showed acute injury and resulted in subsequent development of demyelination, reactive astrocyte, and microglia. Primary effects after IR involved stimulating endothelial cells, increasing dilation and thickening of blood vessels, nuclear enlargement of endothelial cells, and increasing in size and growth of perivascular astrocytes.61 Ionizing radiation could induce over activation of inflammatory cytokines (eg, tumor necrosis factor α [TNF-α]), adhesion molecules (eg, intercellular adhesion molecule 1 [ICAM-1]), chemokine (eg, monocyte chemoattractant protein 1), and matrix metalloproteinases (MMP; eg, MMP-9) that were potential factors of endothelial injury18 and responsible for primary endothelial cell death and apoptosis.62 Cerebral vascular injury was also developed by deteriorating and degenerating structural changes in WM.61 Furthermore, clinical studies showed that radiation-induced WM injury was incorporated with axonal injury, demyelination, neuroinflammation, and necrosis.63-65 In adult rat models, it was reported that after IR regenerative capacity of the oligodendrocyte type 2 astrocyte progenitor cells decreased in both brain and spinal cord causing demyelination.66 Hippocampus was considerably the most radiosensitive and neuroinflammation sensitive area of the brain.67 It was critical to damage the hippocampus which involved the generation of neurons from neural stem cells or progenitor cells throughout the life and cognitive processes such as demonstrative memory and spatial information processing,68 and adverse effects by RT could play a vital role in radiation-induced cognitive impairments. In laboratory mouse models, hippocampus-dependent memory deficiency was observed in adult animals 3 months after cranial irradiation due to decreased hippocampal neurogenesis.69,70 In the same way, after whole-brain irradiation treatment, the numbers of neural stem cells and progenitor cells were declined from the subgranular zone of the hippocampus.56,71

Biomarkers

The term biomarker, a portmanteau of biological marker, generally used in a broad sense, is an indicator or a sign of normal and pathogenic biological processes. As an accusative attribute capable of objectively measuring and accurately evaluating any specific biological and pathological conditions with reproducibility, it has valuable medical applications in diagnosis, staging and prognosis of diseases, monitoring clinical response to therapeutic interventions, and predicting late health consequences. Biomarker is a characteristic, which could be measured reflecting fluctuation of or alteration in any substance, structure, process, or function, and predicting the incidence of outcome or disease, the effects of treatments, interventions, and even unintended environmental exposure. From the point of view on the validity of biomarkers in environment risk assessment, a true definition by World Health Organization of biomarkers includes almost any measurement reflecting an interaction between a biological system and a potential hazard.72-75 For example, a biomarker could be a cell or a molecule in a biological sample collected from the body (ie, complete blood count, circulating DNA, messenger RNA (mRNA), microRNA, and long noncoding RNAs, carcinoembryonic antigens, glucose, proteins, cytokines, growth factors, metabolites in the blood). It could be a result obtained from the imaging technique showing the fluctuation of a substance or alteration in structure and function.76,77 Thus, measurement of a biomarker is not necessarily subject to a biological sample which could be collected; noninvasive diagnostic method, that is, medical imaging techniques using near infrared spectroscopy, is also included.

The biomarker in RT could be grouped into the following categories: (1) the predictive biomarker, available before irradiation, which could predict the outcome and subsequent increased risk of RT, that is, interleukin-1 (IL-1), IL-6, and micronuclei78; (2) the prognostic biomarker, which can be detected at any time after IR and anticipate a consequent increased probability for recurrence or more severe disease, that is, transforming growth factor β1 and fibroblast growth factor 279,80; (3) the diagnostic biomarker, which appears during the symptoms of radiation-induced damage; and (4) the dosimetric biomarker, present at some time point after IR, which is able to determine the radiation dose delivered to the organ. According to the presence of biomarkers in various radiation-induced physiological conditions of the brain, different types of biomarkers are listed in Table 1.

Table 1.

Biomarkers of Brain Damage Induced by Radiotherapy.a

Physiological conditions Biomarkers References
Inflammation Tumor necrosis factor α (TNF-α)
Interleukin (IL-1β, IL-4, IL-6, and IL-8)
Inducible nitric oxide synthase
Intercellular adhesion molecule 1 (ICAM-1)
Matrix metalloproteinase 9 (MMP-9)
Vascular cell adhesion molecule 1
Monocyte chemoattractant protein 1
Cyclooxygenase 1 and 2
9,18,81
Cell activation and damage Glial fibrillary acidic protein
Vascular endothelial growth factor (VEGF)
Vascular cell adhesion molecule
Intercellular adhesion molecule 1 (ICAM-1)
Antiaquaporin-4 antibodies
Neuron-specific enolase
S100 calcium-binding protein B
Myelin basic protein
Cerebrospinal fluid oxysterols
Proteolyzed N-methyl-d-aspartate receptor
Anti-N-methyl-d-aspartate receptor antibodies
Endothelial monocyte-activating
Polypeptide-II cytokine
Nitrotyrosine
14,82-85
Angiogenesis Vascular endothelial growth factor (VEGF)
Angiopoietin (Ang-1, Ang-2, Ang-3, and Ang-4)
Tyrosine-protein kinase (Tie 2)
18,86-89
DNA damage and repair p53-Binding protein 1
Gamma histone protein from the H2A family (γ-H2AX)
Dicentric chromosomes
Micronucleus
(MicroRNA) O6-methylguanine DNA methyltransferase (MGMT)
14,90-93

a Peripheral blood plasma was used to analyze biomarkers for inflammation, cell activation and damage and angiogenesis, and peripheral lymphocytes with complete DNA for DNA damage and repair.

Biomarkers of Radiation-Induced Inflammation

It was identified that oxidative stress and inflammatory pathway were mostly responsible for radiation-induced brain injury.9,81 Significant overexpression of cyclooxygenase 1 and 2 activity and subsequent generation of prostaglandin E2 synthesis lead to developing radiation-induced inflammation in CNS through the upregulatory activity of various proinflammatory mediators including TNF-α, IL-1β, IL-4, IL-6, IL-8, inducible nitric oxide synthase, ICAM-1, and MMP-9. Upregulating adhesion molecules, such as ICAM-1 and vascular cell adhesion molecule 1, was also spotted in radiation-exposed brains. In the hippocampus and the cortical regions, highly overexpressed mRNA encoding cytokines (TNF-α, IL-1β, IL-4, IL-6, IL-8, etc) and protein product of proinflammatory mediators such as TNF-α, IL-1β, and monocyte chemoattractant protein 1 were detected. Area-specific cytokine activation process was observed: TNF-α levels were markedly higher in the cortex than hippocampus, and IL-1β levels were significantly prominent in the hippocampus than the cortical area.18 All of these inflammatory markers were detected in the blood sample of patients after RT.94 These studies suggested that all above-mentioned proinflammatory mediators may be recognized as potential biomarkers of RT-associated damage in CNS.

Biomarkers of Radiation-Induced Cell Activation and Damage

The development of radiation-induced brain injuries became chronic through the activation of various cell subpopulations. Under cerebral pathologic conditions, several biomarkers presented in the peripheral blood indicating specific nerve cell activation or damage. Glial fibrillary acidic protein, vascular endothelial growth factor (VEGF), and vascular cell adhesion molecule were particularly for astrocyte activation, and ICAM-1 was also considered as the endothelium activating biomarker. Antiaquaporin-4 antibodies in blood serum by molecular diagnosis indicated the demyelination of brain cells in patients with tumor after cranial RT.14 Neuron-specific enolase, a neuroendocrine processing glycolytic enzyme, and S100 calcium-binding protein B, a nervous system specific cytoplasmic protein generated by astrocytes when the BBB was ruptured, were regarded as the prospective markers for screening endothelial and neuronal injury. As particular markers, increased neuron-specific enolase denoted brain metastases and predicted shorter survival,82,83 an elevated level of S100 calcium-binding protein B in the circulating blood indicated BBB damage,95 brain metastasis,96 and predicted melanoma brain metastases.97 Demyelination of myelin basic protein was involved with damage of oligodendrocytes.84 Increased cerebrospinal fluid oxysterols in plasma was a promising marker of acute radiation syndrome of CNS.85 Studies also showed that neuron-specific enzyme ubiquitin C-terminal hydrolase, the fragment of proteolytic cleavage of the N-methyl-d-aspartate receptor or N-methyl-d-aspartate receptor antibodies, endothelial monocyte-activating polypeptide-II cytokine, and nitrotyrosine could correlate with brain injury following RT.14 These molecules could be regarded as potential biomarkers and a high level of these molecules in blood serum implied the severity of radiation-induced brain damage.

Biomarkers of Angiogenesis After Irradiation

Angiogenic factors were reported as promising tumor markers in various malignancies.98 For example, VEGF was a key angiogenic factor which could effectively regulate vascular endothelium86-88 involving initiation of endothelial cell proliferation, migration, and propagation of new capillary sprouts, ultimately leading to enhanced vasculogenesis and angiogenesis.99-101 Investigations confirmed that VEGF could promote the growth of tumor cells and protect endothelial cells from apoptosis.88 In addition, all angiopoietin (Ang) families, such as Ang-1, Ang-2, Ang-3, Ang-4, and endothelial cells receptor tyrosine kinase Tie 2 were responsible for the development and integration of endothelial cells; Ang-2 could enhance endothelial cell death and eventually lead to rarefaction of vessels without activating signals from VEGF. Physiological angiogenesis was assisted by Ang-2 through the presence of a high level of VEGF.18 In the rat model for studying endothelial cell proliferation and apoptosis, and expression of various angiogenic factors after whole brain irradiation, it showed that substantially decreased mRNA and protein expression of VEGF, Ang-1, and Tie-2 but significantly upregulated Ang-2 expression were induced,89 suggesting that these angiogenic factors could be used as biomarkers of radiation-induced endothelial cell damage in the brain.

Biomarkers of Radiation-Induced DNA Damage and Repair Processes

Lymphocytes are important cells with complete DNA for screening the effects from exposure to radiation.90 Ionizing radiation could cause various types of chromosomal aberrations, for example, translocations, terminal deletions, ring chromosomes, and dicentric chromosomes.91 Radiation-induced DNA double-strand breaks (DSBs) could activate ataxia telangiectasia mutated (ATM) and ATM- and Rad3-related (ATR) protein kinase, and alter phosphorylation of many cell cycle proteins and DNA repair enzymes,102 leading subsequently to cell cycle arrest or apoptosis.103 Although only a few numbers of lymphocytes were in the brain, lymphocytes collected from peripheral blood with chromosomal aberrations and altered repair proteins accumulated in the area of DSBs, such as p53-binding protein 1 and γ-H2AX involving phosphoinositide 3-kinase and ATM,92,93 could be detected by such as immunofluorescence and flow cytometry microscopy technique for quantitative assessment of IR exposure.104 Studies showed that dicentric chromosomes, micronucleus, and overexpression of some microRNAs including miR-212 could be recognized as valuable biomarkers for both measuring chromosomal damage caused by IR and predicting the progress of radiation injury and outcome for survival.91 Identification of altered repair proteins in the blood with advanced biochemical methods is also an approach to assess the effects of IR on the brain. For example, the repair enzyme O6-methylguanine DNA methyltransferase (MGMT) could enable to protect DNA against alkylating agents (ie, temozolomide) that enhance the risk of radiation-induced injury while transcription of promoter methylation inhibitory enzyme could increase the sensitivity of brain tissue to RT. Therefore, the technique for measurement of MGMT methylation process could determine radiosensitivity and radiation-induced necrosis development.14

Biomarkers of Radiation-Induced Brain Damage Used in Imaging Technology

Radiation-induced detrimental effects generally appeared late and occurred in the closed cranial cavity of the brain,47 making the identification of biomarkers for radiation-induced brain damage, especially the predictive or prognostic ones, more challenging. Novel noninvasive methods are needed to overcome these limitations. Imaging techniques could be considered potential noninvasive biomarkers due to the capability to interrogate metabolic, physiologic, and functional characteristics of the brain and providing significant information for specific areas of normal and tumor tissue.47 The use and validation of both established and new techniques in the context of monitoring early and late brain damage induced by RT in the healthy tissues currently are minimal at best.105,106 In this section, the performance and limitations of existing imaging techniques and the relation of these findings with key clinical parameters were summarized.

Diffusion tensor imaging, as the most sensitive imaging technique, can visualize and evaluate WM integrity and histopathological alterations, distinguish infiltrative growing tumors from bounded tumors and properly specify the tumor grade with conventional magnetic resonance imaging (MRI).107-109 Based on anisotropic diffusion, the value of fractional anisotropy could be used to monitor and detect the early radiation-induced WM injury, that is, alterations in density and orientation of fiber tracts, demyelination or necrosis, and distinguish between demyelination and axonal injury following brain RT.47,110,111 Magnetic resonance imaging is used for the exact screening of tumor, greatly sensitive to pathologic alterations of parenchyma112 but not to low-grade glioma113 and infiltrative tumor growth.114,115 Typical scan includes T1/T2-weighted, fluid-attenuated inversion recovery, and post-contrast T1-weighted images, showing anatomical features, cerebrospinal fluid, pathological conditions, and lesions.112 Functional MRI is mainly to measure blood flow in the region of interest. It could evaluate tumor grade specification, hypoxic, and tumor invasive area by using a T2-weighted signal.116-118 Magnetic resonance imaging can detect radiation-induced acute vascular injury involving blood vessel dilatation, endothelial cell enlargement, capillary loss, astrocyte hypertrophy, BBB disruption, increased permeability, and edema prior to the appearance of radiation-induced demyelination and WM necrosis.119,120 Alteration of BBB permeability after RT, the consequence of endothelial cell damage120 could be monitored with the contrast-enhanced technique using K-trans values.121 Magnetic resonance spectroscopy could provide biochemical and metabolic information of tumors and adjacent tissues evaluate RT-induced necrosis and tumor recurrence, differentiate tumor from lesions122 and recurrent tumor progression from radiation necrosis,123 but less effective to detect the mixed tumor and necrosis.124 Human brain metabolites, such as N-acetyl aspartate, creatine and choline that remained relatively constant125 and were the key marker of the neuronal density and activity, indicator for cellular metabolism and membrane stability, respectively, were most important usable indexes.126 Magnetic resonance spectroscopy could determine the pathological progression process at the biochemical level prior to any conventional techniques.122,125,127 For example, the concentration of N-acetyl aspartate, choline, choline compounds, and choline/creatine ratio significantly decreased after IR, choline/N-acetyl aspartate and choline/creatine ratios are comparatively high in the area of recurrent tumors than the area of radiation injury. Decreased N-acetyl aspartate/creatine ratio indicates neuronal damage, cell death, dysfunction due to apoptosis, and brain irradiation.61 Magnetic resonance spectroscopy was an effective prognostic tool for tumor treatment,128 monitoring response to RT129 and evaluating late-delayed radiation-induced injuries.130 Two-dimensional multivoxel or 3-dimensional spectroscopic imaging technique was used in interval follow-up of RT-treated patients.131 Positron emission tomography (PET) imaging technique could be used in neuro-oncology as an essential tool for grading of primary brain tumors; identifications of neoplastic tissue with delineation of tumor extent for future diagnosis, tumors progression follow-up, and assessment of anticancer treatment response; prognostication; detection of tumor part with a malignant procession; and prediction of biomarkers.132,133 Positron emission tomography can discriminate late-delayed radiation injury from a recurrent brain tumor with 80% to 90% of sensitivity and 50% to 90% of specificity.61 [18F]2-fluoro-2-deoxy-d-glucose was considered as the most important PET modality for the detection of radiation-induced changes in normal brain tissue and neurocognitive impairment.134 With the broad application of radiolabeled amino acid tracers, in addition to significant predicting the outcome of survival,135 PET could precisely differentiate recurrent tumor from radiation-induced necrosis,136 accurately distinguish RT-induced early and late injury from tumor progression.137

Conclusions and Perspectives

Various types of biomarkers have been identified with a variety of measuring techniques in biological and clinical studies. The valuable biomarkers are identified on the basis of some important criteria such as easy collection, instant availability, inexpensiveness, easy detection process, and in particular, of predictive or prognostic ability to specify a disease. Clinical application of a particular and potential biomarker can prevent and mitigate the severity of unbeaten diseases like cancer.

Radiation-induced brain damage is a major dose-limiting adverse event of RT. The incidence varies with the RT modality, dose and its delivery, and the nature of the lesion being targeted and genetic factors of the patients. Biomarkers of brain damage induced by RT are the most objective, quantifiable biological or medical indicators and signs, allowing measurement, evaluation, and prediction of the acute injury and late health consequences with reproducibility. Currently, numerous types of biomarkers are available in the clinical medicine, while biomarkers of radiation-induced brain damage are not available in large quantities. For example, there is no validated biomarker to measure the absorbed dose of the brain after deliberate IR exposure or nuclear accident event.91 As a key issue that innovative approaches to the research, development, and refinement of biomarkers are urgently needed to rapidly advance the research work on the decisive relationship between any biomarker and the relevant clinical consequences, and research and development of the new candidate biomarkers, that is, the endocrine hormones in peripheral blood that have not yet been documented. For imaging biomarkers, though multiple structural and functional imaging modalities exist, each technique independently is not efficient enough, and biochemical indicators combined with imaging techniques will be the development direction of the biomarker application. In addition to the prediction, early detection, and diagnosis of brain injury, research and development of biomarkers could also produce major benefits for monitoring, prognosis, and prediction of therapeutic response of the brain injury, and surveillance of late health consequences after treatment. Although the practical application of biomarkers is moderately new, it is expected that translation of this research advance to RT would assist stratifying patients for optimized treatment, minimizing side effects, and improving therapeutic efficacy and the quality of life.

Acknowledgments

The authors would like to thank Ms Yasuko Morimoto, Ms Hiromi Arai, Ms Mikiko Nakajima, Ms Chianing Hsieh, Ms Kaoru Tanaka, and Dr Masahiro Murakami, Dietary Effects Research Group, Department of Radiation Effects Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Japan, for their kind support throughout the study.

Footnotes

Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work was partially supported by Ministry of Education, Culture, Sport, Sciences and Technology (MEXT) Grant-in-Aid for Scientific Research on Innovative Areas, Grant Number 15H05935 “Living in Space”. The first author was a fellow of the Nuclear Researchers Exchange Programme 2019 supported by MEXT, Japan, and the Nuclear Safety Research Association, Japan.

References

  • 1. Patel S, Bhatnagar A, Wear C, et al. Are pediatric brain tumors on the rise in the USA? Significant incidence and survival findings from the SEER database analysis. Childs Nerv Syst. 2014;30(1):147–154. [DOI] [PubMed] [Google Scholar]
  • 2. Bishop AJ, McDonald MW, Chang AL, Esiashvili N. Infant brain tumors: incidence, survival, and the role of radiation based on Surveillance, Epidemiology, and End Results (SEER) Data. Int J Radiat Oncol Biol Phys. 2012;82(1):341–347. [DOI] [PubMed] [Google Scholar]
  • 3. Armstrong GT. Long-term survivors of childhood central nervous system malignancies: the experience of the Childhood Cancer Survivor Study. Eur J Paediatr Neurol. 2010;14(4):298–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Johnson JD, Young B. Demographics of brain metastasis. Neurosurg Clin N Am. 1996;7(3):337–344. [PubMed] [Google Scholar]
  • 5. Wong CS, Van der Kogel AJ. Mechanisms of radiation injury to the central nervous system: implications for neuroprotection. Mol Interv. 2004;4(5):273–284. [DOI] [PubMed] [Google Scholar]
  • 6. Durand T, Bernier MO, Léger I, et al. Cognitive outcome after radiotherapy in brain tumor. Curr Opin Oncol. 2015;27(6):510–515. [DOI] [PubMed] [Google Scholar]
  • 7. Makale MT, McDonald CR, Hattangadi-Gluth JA, Kesari S. Mechanisms of radiotherapy-associated cognitive disability in patients with brain tumours. Nat Rev Neurol. 2017;13(1):52–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Béhin A, Delattre JY. Complications of radiation therapy on the brain and spinal cord. Semin Neurol. 2004;24(4):405–417. [DOI] [PubMed] [Google Scholar]
  • 9. Denham JW, Hauer-Jensen M. The radiotherapeutic injury—a complex ‘wound’. Radiother Oncol. 2002;63(2):129–145. [DOI] [PubMed] [Google Scholar]
  • 10. Moulder JE, Cohen EP. Future strategies for mitigation and treatment of chronic radiation-induced normal tissue injury. Semin Radiat Oncol. 2007;17(2):141–148. [DOI] [PubMed] [Google Scholar]
  • 11. New P. Radiation injury to the nervous system. Curr Opin Neurol. 2001;14(6):725–734. [DOI] [PubMed] [Google Scholar]
  • 12. Sheline GE, Wara WM, Smith V. Therapeutic irradiation and brain injury. Int J Radiat Oncol Biol Phys. 1980;6(9):1215–1228. [DOI] [PubMed] [Google Scholar]
  • 13. Stone HB, Coleman CN, Anscher MS, McBride WH. Effects of radiation on normal tissue: consequences and mechanisms. Lancet Oncol. 2003;4(9):529–536. [DOI] [PubMed] [Google Scholar]
  • 14. Piskunov AK, Nikitin KV, Potapov AA. Cellular and molecular mechanisms of radiation-induced brain injury: can peripheral markers be detected? Zhurnal voprosy neirokhirurgii imeni N.N. Burdenko. 2015;79(1):90–96. [DOI] [PubMed] [Google Scholar]
  • 15. Biomarkers Definitions Working Group. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther. 2001;69(3):89–95. [DOI] [PubMed] [Google Scholar]
  • 16. Ruberg SJ, Shen L. Personalized medicine: four perspectives of tailored medicine. Stat Biopharm Res. 2015;7(3):214–229. [Google Scholar]
  • 17. Chandana SR, Movva S, Arora M, Singh T. Primary brain tumors in adults. Am Fam Physician. 2008;77(10):1423–1430. [PubMed] [Google Scholar]
  • 18. Lee YW, Cho HJ, Lee WH, Sonntag WE. Whole brain radiation-induced cognitive impairment: pathophysiological mechanisms and therapeutic targets. Biomol Ther. 2012;20(4):357–370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Buckner JC, Brown PD, O’Neill BP, Meyer FB, Wetmore CJ, Uhm JH. Central nervous system tumors. Mayo Clin Proc. 2007;82(10):1271–1286. [DOI] [PubMed] [Google Scholar]
  • 20. Patchell RA, Regine WF. The rationale for adjuvant whole brain radiation therapy with radiosurgery in the treatment of single brain metastases. Technol Cancer Res Treat. 2003;2(2):111–115. [DOI] [PubMed] [Google Scholar]
  • 21. Zimm S, Wampler GL, Stablein D, Hazra T, Young HF. Intracerebral metastases in solid-tumor patients: natural history and results of treatment. Cancer. 1981;48(2):384–394. [DOI] [PubMed] [Google Scholar]
  • 22. Cairncross JG, Kim JH, Posner JB. Radiation therapy for brain metastases. Ann Neurol. 1980;7(3):529–541. [DOI] [PubMed] [Google Scholar]
  • 23. Borgelt B, Gelber R, Kramer S, et al. The palliation of brain metastases: final results of the first two studies by the radiation therapy oncology group. Int J Radiat Oncol Biol Phys. 1980;6(1):1–9. [DOI] [PubMed] [Google Scholar]
  • 24. Lagerwaard FJ, Levendag PC, Nowak PJ, Eijkenboom WM, Hanssens PE, Schmitz PI. Identification of prognostic factors in patients with brain metastases: a review of 1292 patients. Int J Radiat Oncol Biol Phys. 1999;43(4):795–803. [DOI] [PubMed] [Google Scholar]
  • 25. Pannkuk EL, Fornace AJ, Jr, Laiakis EC. Metabolomic applications in radiation biodosimetry. Exploring radiation effects through small molecules. Int J Radiat Biol. 2017;93(10):1151–1176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Kocher M, Soffietti R, Abacioglu U, et al. Adjuvant whole-brain radiotherapy versus observation after radiosurgery or surgical resection of one to three cerebral metastases: Results of the EORTC 22952-26001 study. J Clin Oncol. 2011;29(2):134–141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Chang EL, Wefel JS, Hess KR, et al. Neurocognition in patients with brain metastases treated with radiosurgery or radiosurgery plus whole-brain irradiation: a randomised controlled trial. Lancet Oncol. 2009;10(11):1037–1044. [DOI] [PubMed] [Google Scholar]
  • 28. Bodensohn R, Corradini S, Ganswindt U, et al. A prospective study on neurocognitive effects after primary radiotherapy in high-grade glioma patients. Int J Clin Oncol. 2016;21(4):642–650. [DOI] [PubMed] [Google Scholar]
  • 29. Brown PD, Jaeckle K, Ballman KV, et al. Effect of radiosurgery alone vs radiosurgery with whole brain radiation therapy on cognitive function in patients with 1 to 3 brain metastases: a randomized clinical trial. JAMA. 2016;316(4):401–409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Li J, Bentzen SM, Renschler M, Mehta MP. Regression after whole-brain radiation therapy for brain metastases correlates with survival and improved neurocognitive function. J Clin Oncol. 2007;25(10):1260–1266. [DOI] [PubMed] [Google Scholar]
  • 31. Quik EH, Valk GD, Drent ML, et al. Reduced growth hormone secretion after cranial irradiation contributes to neurocognitive dysfunction. Growth Horm IGF Res. 2012;22(1):42–47. [DOI] [PubMed] [Google Scholar]
  • 32. Sanghavi SN, Miranpuri SS, Chappell R, et al. Radiosurgery for patients with brain metastases: a multi-institutional analysis, stratified by the RTOG recursive partitioning analysis method. Int J Radiat Oncol Biol Phys. 2001;51(2):426–434. [DOI] [PubMed] [Google Scholar]
  • 33. Petrovich Z, Yu C, Giannotta SL, O’Day S, Apuzzo ML. Survival and pattern of failure in brain metastasis treated with stereotactic gamma knife radiosurgery. J Neurosurg. 2002;97(5 suppl):499–506. [DOI] [PubMed] [Google Scholar]
  • 34. Tsao M, Xu W, Sahgal A. A meta-analysis evaluating stereotactic radiosurgery, whole-brain radiotherapy, or both for patients presenting with a limited number of brain metastases. Cancer. 2012;118(9):2486–2493. [DOI] [PubMed] [Google Scholar]
  • 35. Andrews DW, Scott CB, Sperduto PW, et al. Whole brain radiation therapy with or without stereotactic radiosurgery boost for patients with one to three brain metastases: phase III results of the RTOG 9508 randomised trial. Lancet. 2004;363(9422):1665–1672. [DOI] [PubMed] [Google Scholar]
  • 36. Okunieff P, Suit HD. Toxicity, radiation sensitivity modification, and combined drug effects of ascorbic acid with misonidazole in vivo on FSaII murine fibrosarcomas. J Natl Cancer Inst. 1987;79(2):377–381. [PubMed] [Google Scholar]
  • 37. Calvo W, Hopewell JW, Reinhold HS, Yeung TK. Time and dose related changes in the white matter of the rat brain after single doses of X rays. Br J Radiol. 1988;61(731):1043–1052. [DOI] [PubMed] [Google Scholar]
  • 38. Tofilon PJ, Fike JR. The radioresponse of the central nervous system: a dynamic process. Radiat Res. 2000;153(4):357–370. [DOI] [PubMed] [Google Scholar]
  • 39. Sundgren PC, Nagesh V, Elias A, et al. Metabolic alterations: a biomarker for radiation-induced normal brain injury - an MR spectroscopy study. J Magn Reson Imaging. 2009;29 (2):291–297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Shi L, Adams MM, Long A, et al. Spatial learning and memory deficits after whole-brain irradiation are associated with changes in NMDA receptor subunits in the hippocampus. Radiat Res. 2006;166(6):892–899. [DOI] [PubMed] [Google Scholar]
  • 41. Schultheiss TE, Stephens LJC. Permanent radiation myelopathy. Br J Radiol. 1992;65(777):737–753. [DOI] [PubMed] [Google Scholar]
  • 42. Warrington JP, Csiszar A, Mitschelen M, Lee YW, Sonntag WE. Whole brain radiation-induced impairments in learning and memory are time-sensitive and reversible by systemic hypoxia. PLoS One. 2012;7(1):e30444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Johannesen TB, Lien HH, Hole KH, Lote K. Radiological and clinical assessment of long-term brain tumour survivors after radiotherapy. Radiother Oncol. 2003;69(2):169–176. [DOI] [PubMed] [Google Scholar]
  • 44. Meyers CA, Brown PD. Role and relevance of neurocognitive assessment in clinical trials of patients with CNS tumors. J Clin Oncol. 2006;24(8):1305–1309. [DOI] [PubMed] [Google Scholar]
  • 45. Minniti G, De Sanctis V, Muni R, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma in elderly patients. J Neurooncol. 2008;88(1):97–103. [DOI] [PubMed] [Google Scholar]
  • 46. Krex D, Klink B, Hartmann C, et al. Long-term survival with glioblastoma multiforme. Brain. 2007;130(Pt 10):2596–2606. [DOI] [PubMed] [Google Scholar]
  • 47. Greene-Schloesser D, Robbins ME, Peiffer AM, Shaw EG, Wheeler KT, Chan MD. Radiation-induced brain injury: a review. Front Oncol. 2012;2:73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Makale MT, McDonald CR, Hattangadi-Gluth JA, Kesari S. Mechanisms of radiotherapy - associated cognitive disability in patients with brain tumours. Nat Rev Neurol. 2016;13(1):52–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Chapman CH, Nagesh V, Sundgren PC, et al. Diffusion tensor imaging of normal-appearing white matter as biomarker for radiation-induced late delayed cognitive decline. Int J Radiat Oncol Biol Phys. 2012;82(5):2033–2040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Balentova S, Adamkov M. Molecular, cellular and functional effects of radiation-induced brain injury: a review. Int J Mol Sci. 2015;16(11):27796–27815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Belka C, Budach W, Kortmann RD, et al. Radiation induced CNS toxicity—molecular and cellular mechanisms. Br J Cancer. 2001;85(9):1233–1239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Zhao W, Robbins ME. Inflammation and chronic oxidative stress in radiation-induced late normal tissue injury: therapeutic implications. Curr Med Chem. 2009;16(2):130–143. [DOI] [PubMed] [Google Scholar]
  • 53. Furuse M, Nonoguchi N, Kawabata S, et al. Delayed brain radiation necrosis: Pathological review and new molecular targets for treatment. Med Mol Morphol. 2015;48(4):183–190. [DOI] [PubMed] [Google Scholar]
  • 54. Lazarini F, Mouthon MA, Gheusi G, et al. Cellular and behavioral effects of cranial irradiation of the subventricular zone in adult mice. PLoS One. 2009;4(9):e7017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Monje M, Dietrich J. Cognitive side effects of cancer therapy demonstrate a functional role for adult neurogenesis. Behav Brain Res. 2012;227(2):376–379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Pineda JR, Daynac M, Chicheportiche A, et al. Vascular-derived TGF-b increases in the stem cell niche and perturbs neurogenesis during aging and following irradiation in the adult mouse brain. EMBO Mol Med. 2013;5(4):548–562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Farjam R, Pramanik P, Aryal MP, et al. A radiation-induced hippocampal vascular injury surrogate marker predicts late neuro-cognitive dysfunction. Int J Radiat Oncol Biol Phys. 2015;93(4):908–915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Yoritsune E, Furuse M, Kuwabara H, et al. Inflammation as well as angiogenesis may participate in the pathophysiology of brain radiation necrosis. J Radiat Res. 2014;55(4):803–811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Son Y, Yang M, Wang H, et al. Hippocampal dysfunctions caused by cranial irradiation: a review of the experimental evidence. Brain Behav Immun. 2015;45:287–296. [DOI] [PubMed] [Google Scholar]
  • 60. Yang L, Yang J, Li G, et al. Pathophysiological responses in rat and mouse models of radiation-induced brain injury. Mol Neurobiol. 2017;54(2):1022–1032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Sundgren PC, Cao Y. Brain irradiation: effects on normal brain parenchyma and radiation injury. Neuroimaging Clin N Am. 2009;19(4):657–668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Santana P, Pena LA, Haimovitz-Friedman A, et al. Acid sphingomyelinase- deficient human lymphoblasts and mice are defective in radiation-induced apoptosis. Cell. 1996;86(2):189–199. [DOI] [PubMed] [Google Scholar]
  • 63. Leeds NE, Fuller GN, Van Tassel P, et al. Malignant gliomas: MR imaging spectrum of radiation therapy and chemotherapy-induced necrosis of the brain after treatment. Radiology. 2000;217(2):377–384. [DOI] [PubMed] [Google Scholar]
  • 64. Nagesh V, Tsien CI, Chenevert TL, et al. Radiation-induced changes in normal-appearing white matter in patients with cerebral tumors: a diffusion tensor imaging study. Int J Radiat Oncol Biol Phys. 2008;70(4):1002–1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Wang S, Wu EX, Qiu D, Leung LH, Lau HF, Khong PL. Longitudinal diffusion tensor magnetic resonance imaging study of radiation-induced white matter damage in a rat model. Cancer Res. 2009;69(3):1190–1198. [DOI] [PubMed] [Google Scholar]
  • 66. van der Maazen RW, Kleiboer BJ, Verhagen I, et al. Irradiation in vitro discriminates between different O-2A progenitor cell subpopulations in the perinatal central nervous system of rats. Radiat Res. 1991;128(1):64–672. [PubMed] [Google Scholar]
  • 67. Piskunov AK, Yakovlev AA, Stepanichev MY, Onufriev MV, Gulyaeva NV. Selective vulnerability of the hippocampus to interoceptive stress: effects on interleukin-1β and erythropoietin. Neurochem J. 2011;5(3):191–193. [Google Scholar]
  • 68. Eichenbaum H, Dudchenko P, Wood E, et al. The hippocampus, memory, and place cells: is it spatial memory or a memory space? Neuron. 1999;23(2):209–226. [DOI] [PubMed] [Google Scholar]
  • 69. Rola R, Raber J, Rizk A, et al. Radiation-induced impairment of hippocampal neurogenesis is associated with cognitive deficits in young mice. Exp Neurol. 2004;188(2):316–330. [DOI] [PubMed] [Google Scholar]
  • 70. Raber J, Rola R, LeFevour A, et al. Radiation-induced cognitive impairments are associated with changes in indicators of hippocampal neurogenesis. Radiat Res. 2004;162(1):39–47. [DOI] [PubMed] [Google Scholar]
  • 71. Amano T, Inamura T, Wu CM, et al. Effects of single low dose irradiation on subventricular zone cells in juvenile rat brain. Neurol Res. 2002;24(8):809–816. [DOI] [PubMed] [Google Scholar]
  • 72. WHO International Programme on Chemical Safety Biomarkers and Risk Assessment: Concepts and Principles. 1993. http://www.inchem.org/documents/ehc/ehc/ehc155.htm
  • 73. WHO International Programme on Chemical Safety Biomarkers in Risk Assessment: Validity and Validation. 2001. http://www.inchem.org/documents/ehc/ehc/ehc222.htm
  • 74. Atkinson AJ, Jr, Warren G, Colburn WA, et al. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther. 2001;69(3):89–95. [DOI] [PubMed] [Google Scholar]
  • 75. Strimbu K, Tavel JA. What are biomarkers? Curr Opin HIV AIDS. 2010;5(6):463–466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Sproull M, Camphausen K. State-of-the-art advances in radiation biodosimetry for mass casualty events involving radiation exposure. Radiat Res. 2016;186(5):423–435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Singh VK, Newman VL, Romaine PL, Hauer-Jensen M, Pollard HB. Use of biomarkers for assessing radiation injury and efficacy of countermeasures. Expert Rev Mol Diagn. 2016;16(1):65–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Chen Y, Williams J, Ding I, et al. Radiation pneumonitis and early circulatory cytokine markers. Semin Radiat Oncol. 2002;12(1 suppl 1):26–33. [DOI] [PubMed] [Google Scholar]
  • 79. Fleckenstein K, Gauter-Fleckenstein B, Jackson IL, Rabbani Z, Anscher M, Vujaskovic Z. Using biological markers to predict risk of radiation injury. Semin Radiat Oncol. 2007;17(2):89–98. [DOI] [PubMed] [Google Scholar]
  • 80. Andreassen CN, Alsner J, Overgaard M, Sorensen FB, Overgaard J. Risk of radiation-induced subcutaneous fibrosis in relation to single nucleotide polymorphisms in TGFB1, SOD2, XRCC1, XRCC3, APEX and ATM--a study based on DNA from formalin fixed paraffin embedded tissue samples. Int J Radiat Biol. 2006;82(8):577–586. [DOI] [PubMed] [Google Scholar]
  • 81. Baluna RG, Eng TY, Thomas CR. Adhesion molecules in radiotherapy. Radiat Res. 2006;166(6):819–831. [DOI] [PubMed] [Google Scholar]
  • 82. van de Pol M, Twijnstra A, ten Velde GP, Menheere PP. Neuron-specific enolase as a marker of brain metastasis in patients with small-cell lung carcinoma. J Neurooncol. 1994;19(2):149–154. [DOI] [PubMed] [Google Scholar]
  • 83. Jacot W, Quantin X, Boher JM, et al. Brain metastases at the time of presentation of non-small cell lung cancer: a multicentric AERIO analysis of prognostic factors. Br J Cancer. 2001;84(7):903–909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Montague P, Mccallion AS, Davies RW, Griffiths IR. Myelin-associated oligodendrocytic basic protein: a family of abundant CNS myelin proteins in search of a function. Dev Neurosci. 2006;28(6):479–487. [DOI] [PubMed] [Google Scholar]
  • 85. Bertho JM, Roy L, Souidi M, et al. Initial evaluation and follow-up of acute radiation syndrome in two patients from the Dakar accident. Biomarkers. 2009;14(2):94–102. [DOI] [PubMed] [Google Scholar]
  • 86. Ferrara N. Vascular endothelial growth factor: molecular and biological aspects. Curr Top Microbiol Immunol.1999;237:1–30. [DOI] [PubMed] [Google Scholar]
  • 87. Ribatti D. The crucial role of vascular permeability factor/vascular endothelial growth factor in angiogenesis: a historical review. Br J Haematol. 2005;128(3):303–309. [DOI] [PubMed] [Google Scholar]
  • 88. Tammela T, Enholm B, Alitalo K, Paavonen K. The biology of vascular endothelial growth factors. Cardiovasc Res. 2005;65(3):550–563. [DOI] [PubMed] [Google Scholar]
  • 89. Lee WH, Cho HJ, Sonntag WE, Lee YW. Radiation attenuates physiological angiogenesis by differential expression of VEGF, Ang-1, tie-2 and Ang-2 in rat brain. Radiat Res. 2011;176(6):753–760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Chua ML, Rothkamm K. Biomarkers of radiation exposure: can they predict normal tissue radiosensitivity? Clin Oncol. 2013;25(10):610–616. [DOI] [PubMed] [Google Scholar]
  • 91. Singh VK, Santiago PT, Simas M, et al. Acute radiation syndrome: an update on biomarkers for radiation injury. J Radiat Cancer Res. 2018;9(4):132–146. [Google Scholar]
  • 92. Lord CJ, Garrett MD, Ashworth A. Targeting the double-strand DNA break repair pathway as a therapeutic strategy. Clin Cancer Res. 2006;12:(15):4463–4468. [DOI] [PubMed] [Google Scholar]
  • 93. Kuo LJ, Yang LX. Gamma-H2AX – a novel biomarker for DNA double-strand breaks. In vivo. 2008;22(3):305–309. [PubMed] [Google Scholar]
  • 94. Wickremesekera JK, Chen W, Cannan RJ, Stubbs RS. Serum pro-inflammatory cytokine response in patients with advanced liver tumors following selective internal radiation therapy (SIRT) with (90) Yttrium microspheres. Int J Radiat Oncol Biol Phys. 2001;49(4):1015–1021. [DOI] [PubMed] [Google Scholar]
  • 95. Foerch C, du Mesnil de Rochemont R, Singer O, et al. S100B as a surrogate marker for successful clot lysis in hyperacute middle cerebral artery occlusion. J Neurol Neurosurg Psychiatry. 2003;74(3):322–325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Vogelbaum MA, Masaryk T, Mazzone P, et al. S100beta as a predictor of brain metastases: brain versus cerebrovascular damage. Cancer. 2005;104(4):817–824. [DOI] [PubMed] [Google Scholar]
  • 97. Kaskel P, Berking C, Sander S, Volkenandt M, Peter RU, Krahn G. S-100 protein in peripheral blood: a marker for melanoma metastases: a prospective 2-center study of 570 patients with melanoma. J Am Acad Dermatol. 1999;41(6):962–969. [DOI] [PubMed] [Google Scholar]
  • 98. Chan LW, Moses MA, Goley E, et al. Urinary VEGF and MMP levels as predictive markers of 1-year progression-free survival in cancer patients treated with radiation therapy: a longitudinal study of protein kinetics throughout tumor progression and therapy. J Clin Oncol. 2004;22(3):499–506. [DOI] [PubMed] [Google Scholar]
  • 99. Breier G, Albrecht U, Sterrer S, Risau W. Expression of vascular endothelial growth factor during embryonic angiogenesis and endothelial cell differentiation. Development. 1992;114(2):521–532. [DOI] [PubMed] [Google Scholar]
  • 100. Plate KH. Mechanisms of angiogenesis in the brain. J Neuropathol Exp Neurol. 1999;58(4):313–320. [DOI] [PubMed] [Google Scholar]
  • 101. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med. 2003;9(6):669–676. [DOI] [PubMed] [Google Scholar]
  • 102. Jazayeri A, Falck J, Lukas C, et al. ATM- and cell cycle-dependent regulation of ATR in response to DNA double-strand breaks. Nature Cell Biol. 2006;8(1):37–45. [DOI] [PubMed] [Google Scholar]
  • 103. Vallard A, Rancoule C, Guy JB, et al. Biomarkers of radiation-induced DNA repair processes. Bull Cancer. 2017;104(11):981–987. [DOI] [PubMed] [Google Scholar]
  • 104. Hall J, Jeggo PA, West C, et al. Ionizing radiation biomarkers in epidemiological studies - an update. Mutat Res. 2017;771:59–84. [DOI] [PubMed] [Google Scholar]
  • 105. Ali FS, Arevalo O, Zorofchian S, et al. Cerebral radiation necrosis: Incidence, pathogenesis, diagnostic challenges, and future opportunities. Curr Oncol Rep. 2019;21(8):66. [DOI] [PubMed] [Google Scholar]
  • 106. Kłos J, van Laar PJ, Sinnige PF, et al. Quantifying effects of radiotherapy-induced microvascular injury; review of established and emerging brain MRI techniques. Radiother Oncol. 2019;140:41–53. [DOI] [PubMed] [Google Scholar]
  • 107. Mori S, Crain BJ, Chacko VP, Van Zijl PC. Three-dimensional tracking of axonal projections in the brain by magnetic resonance imaging. Ann Neurol. 1999;45(2):265–269. [DOI] [PubMed] [Google Scholar]
  • 108. Kono K, Inoue Y, Nakayama K, et al. The role of diffusion-weighted imaging in patients with brain tumors. Am J Neuroradiol. 2001;22(6):1081–1088. [PMC free article] [PubMed] [Google Scholar]
  • 109. Kleiser R, Staempfli P, Valavanis A, Boesiger P, Kollias S. Impact of fMRI-guided advanced DTI fiber tracking techniques on their clinical applications in patients with brain tumors. Neuroradiology. 2010;52(1):37–46. [DOI] [PubMed] [Google Scholar]
  • 110. Romano A, Fasoli F, Ferrante M, Ferrante L, Fantozzi LM, Bozzao A. Fiber density index, fractional anisotropy, ADC and clinical motor findings in the white mater of patients with glioblastoma. Eur Radiol. 2008;18(2):331–336. [DOI] [PubMed] [Google Scholar]
  • 111. Basser PJ, Pierpaoli C. A simplified method to measure the diffusion tensor from seven MR images. Magn Reson Med. 1998;39(6):928–934. [DOI] [PubMed] [Google Scholar]
  • 112. Rees J. Advances in magnetic resonance imaging of brain tumours. Curr Opin Neurol. 2003;16(6):643–650. [DOI] [PubMed] [Google Scholar]
  • 113. Pallud J, Capelle L, Taillandier L. Prognostic significance of imaging contrast enhancement for WHO grade II gliomas. Neuro Oncol. 2009;11(2):176–182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Kickingereder P, Götz M, Muschelli J, et al. Large-scale radiomic profiling of recurrent glioblastoma identifies an imaging predictor for stratifying anti-angiogenic treatment response. Clin Cancer Res. 2016;22(23):5765–5771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Zhou M, Chaudhury B, Hall LO, et al. Identifying spatial imaging biomarkers of glioblastoma multiforme for survival group prediction. J Magn Reson Imaging. 2015;42(5):1421–1430. [DOI] [PubMed] [Google Scholar]
  • 116. Wang L, Chen D, Olson J, Ali S, Fan T, Mao H. Re-examine tumor-induced alterations in hemodynamic responses of BOLD fMRI: implications in presurgical brain mapping. Acta Radiol. 2012;53(7):802–811. [DOI] [PubMed] [Google Scholar]
  • 117. Hou BL, Bradbury M, Peck KK, Petrovich NM, Gutin PH, Holodny AI. Effect of brain tumor neovasculature defined by rCBV on BOLD fMRI activation volume in the primary motor cortex. NeuroImage. 2006;32(2):489–497. [DOI] [PubMed] [Google Scholar]
  • 118. Brindle KM, Izquierdo-García JL, Lewis DY, Mair RJ, Wright AJ. Brain tumor imaging. J Clin Oncol. 2017;35(21):2432–2438. [DOI] [PubMed] [Google Scholar]
  • 119. Reinhold HS, Calvo W, Hopewell JW, van den Berg AP. Development of blood vessel-related radiation damage in the fimbria of the central nervous system. Int J Radiat Oncol Biol Phys.1990;18(1):37–42. [DOI] [PubMed] [Google Scholar]
  • 120. Li YQ, Chen P, Haimovitz-Friedman A, Reilly RM, Wong CS. Endothelial apoptosis initiates acute blood–brain barrier disruption after ionizing radiation. Cancer Res. 2003;63(18):5950–5956. [PubMed] [Google Scholar]
  • 121. Tofts PS, Brix G, Buckley DL, et al. Estimating kinetic parameters from dynamic contrast-enhanced T (1)-weighted MRI of a diffusable tracer: standardized quantities and symbols. J Magn Reson Imaging. 1999;10(3):223–232. [DOI] [PubMed] [Google Scholar]
  • 122. Schlemmer HP, Bachert P, Henze H, et al. Differentiation of radiation necrosis from tumor progression using proton magnetic resonance spectroscopy. Neuroradiology. 2002;44(3):216–222. [DOI] [PubMed] [Google Scholar]
  • 123. Chong VF, Khoo JB, Chan LL, Rumpel H. Neurological changes following radiation therapy for head and neck tumours. Eur J Radiol. 2002;44(2):120–129. [DOI] [PubMed] [Google Scholar]
  • 124. Rock JP, Scarpace L, Hearshen D, et al. Associations among magnetic resonance spectroscopy, apparent diffusion coefficients and image-guided histopathology with special attention to radiation necrosis. Neurosurgery. 2004;54(5):1111–1119. [DOI] [PubMed] [Google Scholar]
  • 125. Howe FA, Barton SJ, Cudlip SA, et al. Metabolic profiles of human brain tumors using quantitative in vivo 1 H magnetic resonance spectroscopy. Magn Reson Med. 2003;49(2):223–232. [DOI] [PubMed] [Google Scholar]
  • 126. Villanueva-Meyer JE, Mabray MC, Cha S. Current clinical brain tumor imaging. Neurosurgery. 2017;81(3):397–415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Majós C, Aguilera C, Cos M, et al. In vivo proton magnetic resonance spectroscopy of intraventricular tumors of the brain. Eur Radiol. 2009;19(8):2049–2059. [DOI] [PubMed] [Google Scholar]
  • 128. Alexander A, Murtha A, Abdulkarim B, et al. Prognostic significance of serial magnetic resonance spectroscopies over the course of radiation therapy for patients with malignant glioma. Clin Invest Med. 2006;29(5):301–311. [PubMed] [Google Scholar]
  • 129. Davidson A, Tait DM, Payne GS, et al. Magnetic resonance spectroscopy in the evaluation of neurotoxicity following cranial irradiation for childhood cancer. Br J Radiol. 2000;73(868):421–424. [DOI] [PubMed] [Google Scholar]
  • 130. Usenius T, Usenius JP, Tenhunen M, et al. Radiation-induced changes in human brain metabolites as studied by 1H nuclear magnetic resonance spectroscopy in vivo. Int J Radiat Oncol Biol Phys. 1995;33(3):719–724. [DOI] [PubMed] [Google Scholar]
  • 131. Lee MC, Pirzkall A, McKnight TR, et al. 1H-MRS of radiation effects in normal-appearing white matter: dose-dependence and impact on automated spectral classiftcation. J Magn Reson Imaging. 2004;19(4):379–388. [DOI] [PubMed] [Google Scholar]
  • 132. Albert NL, Weller M, Suchorska B, et al. Response assessment in neurooncology working group and European association for neuro-oncology recommendations for the clinical use of PET imaging in gliomas. Neuro Oncol. 2016;18(9):1199–1208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Galldiks N, Lohmann P, Albert NL, Tonn JC, Langen KJ. Current status of PET imaging in neuro-oncology. Neuro-Oncol Adva. 2019;20(20):1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Herholz K, Langen KJ, Schiepers C, Mountz JM. Brain tumors. Semin Nucl Med. 2012;42(6):356–370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Galldiks N, Dunkl V, Ceccon G, et al. Early treatment response evaluation using FET PET compared to MRI in glioblastoma patients at first progression treated with bevacizumab plus lomustine. Eur J Nucl Med Mol Imaging. 2018;45(13):2377–2386. [DOI] [PubMed] [Google Scholar]
  • 136. Langen KJ, Stoffels G, Filss C, et al. Imaging of amino acid transport in brain tumours: positron emission tomography with O-(2-[18F] fluoroethyl)-L-tyrosine (FET). Methods. 2017;130:124–134. [DOI] [PubMed] [Google Scholar]
  • 137. Mihovilovic MI, Kertels O, Hänscheid H, et al. O-(2-(18F) fluoroethyl)- L-tyrosine PET for the differentiation of tumour recurrence from late pseudoprogression in glioblastoma. J Neurol Neurosurg Psychiatry. 2019;90(2):238–239. [DOI] [PubMed] [Google Scholar]

Articles from Dose-Response are provided here courtesy of SAGE Publications

RESOURCES