v
Search
Advanced

Publications > Journals > Neurosurgical Subspecialties> Article Full Text

  • OPEN ACCESS

State-of-the-art Multimodal Imaging and Therapeutic Strategies in Radiation-induced Brain Injury: A Comprehensive Review

  • Xiaojie Wang,
  • Shuang Li,
  • Fangjing Yu and
  • Xiaonan Cui* 
 Author information 

Abstract

Radiotherapy remains one of the essential treatment modalities for brain gliomas, brain metastases, pediatric neuroblastomas, and primary central nervous system lymphomas. With continuous advancements in modern radiotherapy techniques, patients have achieved significantly improved local control rates and prolonged survival. However, the long-term complications associated with radiotherapy have become increasingly evident. Radiation-induced brain injury (RIBI) is a clinical syndrome characterized primarily by neurological dysfunction following focal or whole-brain radiotherapy. It negatively impacts patients’ quality of life and imposes a considerable burden on families and society. With the rapid development of medical imaging and artificial intelligence technologies, multimodal imaging techniques, including structural magnetic resonance imaging, diffusion-weighted imaging, functional magnetic resonance imaging, perfusion imaging, positron emission tomography-computed tomography metabolic imaging, and radiomics, have demonstrated significant potential for early detection, dynamic monitoring, and quantitative evaluation of RIBI. Meanwhile, treatment strategies for RIBI are shifting from traditional symptomatic and supportive care toward multidimensional interventions aimed at protecting the blood-brain barrier, modulating neuroinflammation, and implementing precise targeted therapies. Additionally, emerging studies have explored neuromodulation techniques and gut-brain axis regulation, offering new directions for the prevention and treatment of RIBI. Although conventional imaging methods remain valuable for diagnosing RIBI, they exhibit notable limitations in the early stages of the disease and in differentiating RIBI from tumor recurrence. This review focuses on the current state of technological development, key findings, and existing limitations, with the aim of providing a theoretical foundation and technical support for the early identification and precise intervention of RIBI.

Keywords

Radiotherapy, Radiation injuries, Brain injuries, Magnetic resonance imaging, Diffusion magnetic resonance imaging, Radiomics, Artificial intelligence, Neuroinflammation

Introduction

Radiation-induced brain injury (RIBI) is a significant complication following cranial radiotherapy, characterized by central nervous system dysfunction. While advances in radiotherapy techniques and systemic therapies have substantially improved overall survival in brain tumor patients, these gains have been offset by a rising incidence of RIBI, which adversely affects survivors’ quality of life.1

Early clinical observations highlighted the harmful effects of nuclear radiation on the developing brain.2 Subsequent research has confirmed that cranial radiotherapy increases the risk of learning difficulties, mood disturbances, and psychiatric disorders.3,4 Although preclinical studies suggest that early intervention may mitigate RIBI symptoms, no universally accepted strategy exists for preventing or reversing the condition. Consequently, the early diagnosis, prevention, and management of RIBI have become key areas of current research.

This review provides a comprehensive summary of recent advances in multimodal imaging diagnostics, including structural magnetic resonance imaging (MRI), diffusion/perfusion modalities, magnetic resonance spectroscopy (MRS)/positron emission tomography (PET), and radiomics, as well as evolving therapeutic strategies for RIBI. Our analysis critically examines the current strengths and limitations of these approaches, identifies gaps in the evidence, and proposes future directions to support early detection and precision management of RIBI in clinical practice. To conduct this review, we searched PubMed, Embase, and Web of Science for studies published up to July 2025, using combinations of keywords such as radiation-induced brain injury, multimodal imaging, MRI, PET, radiomics, and therapeutic strategies. We included peer-reviewed original studies and reviews addressing RIBI pathophysiology, diagnostic imaging, or interventions, while excluding non-English articles, case reports, and publications lacking substantive data.

Pathophysiological mechanisms of RIBI

RIBI is a multifactorial, delayed-onset neurological disorder.5 Its pathogenesis involves a confluence of physiological and pathological processes, including neuronal damage, blood-brain barrier (BBB) disruption, inflammatory responses, cerebrovascular alterations, and aberrant activation of glial cells (Fig. 1).6–9

Mechanisms of radiation-induced brain injury at different post-radiotherapy time points.
Fig. 1  Mechanisms of radiation-induced brain injury at different post-radiotherapy time points.

Radiotherapy induces DNA damage and oxidative stress, leading to endothelial injury and blood-brain barrier disruption, which triggers neuroinflammation, microvascular dysfunction, and neuronal apoptosis, ultimately resulting in progressive edema, demyelination, and irreversible brain necrosis. Created with BioRender.com. ROS, reactive oxygen species.

Blood-brain barrier disruption and endothelial cell injury

Ionizing radiation directly damages cerebral microvascular endothelial cells, leading to increased BBB permeability. Vascular injury can manifest within 24 h post-radiotherapy. This disruption facilitates the extravasation of plasma proteins and the infiltration of activated leukocytes and inflammatory mediators into the central nervous system, exacerbating local inflammation. Early imaging and pathological studies have established a correlation between vascular pathology and cognitive deficits.10–12 Autopsy findings in patients with delayed RIBI reveal widespread vascular abnormalities and demyelination of cerebral white matter.13 Radiation has been shown to activate the phosphatidylinositol 3-kinase – protein kinase B (PI3K-AKT) pathway in astrocytes, reducing autophagy and leading to over-secretion of vascular endothelial growth factor, which contributes to BBB damage.14

Radiotherapy also leads to intracranial vascular occlusion, characterized by shortened vessel length and reduced capillary density. Brown et al.15 observed significant reductions in vascular density and length in irradiated rat brains, correlating with cognitive decline over time. Radiation-induced damage to vascular endothelial cells, coupled with reductions in vascular density and length, compromises BBB integrity, representing an early stage of RIBI.

Inflammatory cytokines and biomarkers of RIBI

RIBI is a complex process involving the interplay of various inflammatory and cellular cytokines. Studies have shown that radiotherapy activates N-methyl-D-aspartate receptors, increasing intracellular calcium levels and activating calcineurin. The nuclear factor of activated T-cells (NFAT3/c4), a downstream target of calcineurin, translocates to the nucleus and promotes transcriptional activity, resulting in neuronal apoptosis.16 Additionally, activation of the kynurenine pathway has been associated with RIBI. Following whole-brain radiotherapy, the kynurenine pathway is significantly activated, characterized by decreased tryptophan levels and increased activity of indoleamine 2,3-dioxygenase and tryptophan 2,3-dioxygenase. Elevated levels of kynurenic acid and quinolinic acid can induce oxidative stress, inflammation, nitric oxide pathway dysregulation, and neuronal apoptosis.17 Moreover, interleukin (IL)-6 and IL-1β are markedly upregulated in brain tissue following radiotherapy,18 and IL-6 has been shown to exacerbate radiation-induced cellular senescence in animal models.19 The release of these inflammatory cytokines and activation of inflammatory pathways may damage neural cells, contributing to cognitive deficits.

Neuronal apoptosis and glial cell activation

Radiation induces cell death through both direct and indirect mechanisms. Direct damage involves DNA strand breaks caused by radiation, leading to cell death.20 Indirect damage occurs via the generation of reactive oxygen species, resulting in DNA damage, impaired protein synthesis, and cell cycle arrest. This cascade leads to cellular swelling, necrosis, and further the production of reactive oxygen species, perpetuating inflammation and increased capillary permeability, ultimately culminating in irreversible radiation-induced brain necrosis.21 These direct and indirect forms of damage reduce neural cell numbers, contributing to brain dysfunction within irradiated regions.

White matter injury and demyelination

White matter regions are rich in glial cells and neurons. Glial cells are more sensitive to radiation than neurons, and their gradual loss within white matter is considered a fundamental aspect of RIBI development.22 As primary responders to central nervous system injury, microglia may promote astrocyte senescence. Research indicates that astrocytes preferentially undergo senescence after radiotherapy, while neural progenitor cells are more prone to apoptosis, suggesting that the secretory phenotype of senescent astrocytes may underlie chronic RIBI.23 Radiation-induced white matter pathology disrupts neural signal transmission, leading to cognitive impairment and neurodegeneration.

Genetic susceptibility and RIBI

Beyond radiation and tumor-specific factors, individual radiosensitivity varies among patients. A genome-wide association study including nasopharyngeal carcinoma patients identified polymorphisms in the CEP128 promoter—such as rs17111237 and rs162171—as significantly associated with differential risk of temporal lobe radiation-induced injury (hazard ratios ∼1.45–1.46, P < 10−7).24 This represents the first study implicating CEP128 in RIBI, offering new insights into potential mechanisms.

Treatment-related factors

RIBI risk is influenced by radiotherapy modality, total dose, fractionation schemes, and the concurrent use of chemotherapy, targeted therapy, or immunotherapy. Advances in techniques such as intensity-modulated radiotherapy, stereotactic radiosurgery (SRS), and image-guided radiotherapy have improved precision, reduced exposure to surrounding normal tissues, and decreased RIBI risk.25

Studies from the Radiation Therapy Oncology Group have shown a positive correlation between RIBI risk and total radiation dose.26 For example, in patients with high-grade gliomas treated with SRS, the incidence of RIBI is approximately 6.5%. When total doses range between 18 and 30 Gy, incidence varies from 4.7% to 9.2%, with higher doses associated with greater risk.

Tumor size also influences RIBI risk, with larger tumors correlating with higher incidence following SRS (2.9–22.6%).27 Radiation-induced cognitive impairment is related to the structure and dose thresholds of irradiated targets. Localized brain irradiation may not cause cognitive deficits of the same magnitude as whole-brain irradiation.28,29 Selective damage to critical brain structures, such as the hippocampus and temporal lobes, may underlie post-radiotherapy cognitive dysfunction; preserving these regions can help maintain cognitive function.

The impact of combined therapies on RIBI remains under investigation. While some retrospective studies suggest agents such as bevacizumab and immune checkpoint inhibitors do not increase RIBI incidence, further research is needed to validate these findings.30,31 With continued advances in radiotherapy techniques, improved treatment planning, and the development of pharmacological interventions, the incidence of RIBI is expected to further decline.

Clinical manifestations of RIBI

RIBI is typically categorized into three phases based on onset time and pathological progression: acute, subacute (early-delayed), and late-delayed phases.32

Acute phase (within hours to weeks post-radiotherapy)

This phase is primarily characterized by symptoms indicative of increased intracranial pressure, including headache, nausea, vomiting, and somnolence. These manifestations are often attributed to reversible cerebral edema and are relatively uncommon in clinical practice. The underlying pathophysiology involves disruption of the BBB and alterations in capillary permeability, leading to vasogenic edema.

Subacute phase (one to six months post-radiotherapy)

During this phase, patients may experience transient cognitive impairments, somnolence, emotional fluctuations, and mild memory decline. Neuroimaging frequently reveals diffuse white matter signal changes. While some cases resolve spontaneously, others may progress to late-delayed brain injury. The pathogenesis is thought to involve transient demyelination and oligodendrocyte dysfunction, potentially linked to BBB disruption.

Late-delayed phase (beyond six months post-radiotherapy)

This phase represents the most common and severe form of RIBI. Clinical features include progressive cognitive decline, disorientation, language and executive function deficits, and, in severe cases, seizures, psychiatric disturbances, coma, or death. Neuroimaging findings often demonstrate brain atrophy, necrotic lesions, and progressive white matter changes. The underlying mechanisms are multifactorial, involving vascular abnormalities, demyelination, and white matter necrosis.

The risk of developing RIBI is influenced by various factors, including radiation dose and fractionation schedule, irradiated brain volume, patient age, comorbid conditions (such as hypertension and diabetes), and concurrent systemic chemotherapy. Accurate recognition of the clinical features and an understanding of the pathophysiological mechanisms are crucial for early detection, differential diagnosis, and precise management of RIBI.

Advances in multimodal imaging for the diagnosis of RIBI

Advanced MRI technology

RIBI is challenging to detect early due to its insidious onset and non-specific clinical symptoms. Moreover, distinguishing its imaging features from tumor recurrence remains difficult using a single modality. In recent years, multimodal imaging strategies centered on MRI, integrating structural, functional, metabolic, perfusion, and artificial intelligence analyses, have emerged as crucial approaches for the precise assessment of RIBI (Table 1).

Table 1

Comparison of multimodal imaging technologies

Multimodal imagingPrincipleApplicationsPros and Cons
FLAIRSuppressing cerebrospinal fluid (CSF) signal to highlight abnormal tissueDetecting parenchymal abnormalitiesHighly sensitive to white matter demyelination
DWIMeasuring water molecular diffusionDifferentiating hypercellular tumors, radiation necrosisProvides apparent diffusion coefficient (ADC) values
DTIAnalyzing the diffusion of water molecules along axonal fibersAssessing white matter microstructureUseful for detecting demyelination and axonal loss; Limitation in crossing fiber evaluation
PWIEvaluating cerebral perfusionMonitoring cerebral perfusionNeed for contrast agents: Sensitivity to artifacts. Contrast agents; Longer scan time
MRSQuantifying brain metabolite concentrationQuantifying radiation-induced neurotoxicityProvides non-invasive functional imaging
BOLDBased on blood oxygenation level-dependentAssessing brain function and hemodynamicProvides non-invasive functional imaging

Within MRI sequences, T1-weighted imaging, T2-weighted imaging, and fluid-attenuated inversion recovery (FLAIR) are highly sensitive in detecting parenchymal abnormalities, white matter lesions, necrotic foci, and brain atrophy. FLAIR is particularly sensitive to white matter demyelination, often presenting as hyperintense areas within or at the margins of the irradiated field.33

Diffusion imaging techniques reflect the diffusion state of water molecules within tissues. Diffusion-weighted imaging (DWI) can evaluate cellular density and regions with restricted water diffusion, aiding in differentiating hypercellular tumor areas from necrotic tissue. DWI provides apparent diffusion coefficient (ADC) values, also referred to as mean diffusivity. Rapid tumor cell proliferation increases cellular density in lesions, leading to decreased ADC values. In contrast, neuronal necrosis following RIBI reduces cellular density, resulting in increased ADC values.34 Diffusion imaging enables the assessment of microstructural changes in brain tissue. Integration of relative cerebral blood volume (CBV) and Ktrans has achieved diagnostic accuracy exceeding 90% in glioblastoma follow-up.35

Diffusion tensor imaging offers additional parameters, including fractional anisotropy,36,37 axial diffusivity, and radial diffusivity. Several studies have focused on microstructural changes in white matter following radiotherapy, showing decreased fractional anisotropy, increased mean diffusivity,38,39 and elevated radial diffusivity,38,40,41 primarily attributed to demyelination or axonal loss.42,43 Perfusion imaging techniques, by measuring cerebral blood flow and CBV, help differentiate pathological conditions. Studies have shown that regions of radiation necrosis often exhibit hypoperfusion (low cerebral blood flow/CBV), whereas tumor recurrence typically shows hyperperfusion. Among these techniques, arterial spin labeling (ASL) does not require contrast agents and is suitable for repeated monitoring, while dynamic contrast-enhanced (DCE) and dynamic susceptibility contrast (DSC) perfusion imaging provide vascular permeability and perfusion curves, offering high sensitivity in assessing post-radiotherapy lesion activity. Perfusion-weighted imaging (PWI) can detect radiation-induced vascular changes; most PWI studies report reduced perfusion and decreased CBV following radiotherapy.44,45 A combination of PWI and DWI improves diagnostic performance and can obviate unnecessary biopsies in approximately 10% of cases.46 Common MRI perfusion techniques in clinical practice include DSC, DCE, and ASL, each with specific advantages and limitations. DSC, based on T2-weighted imaging following rapid contrast injection, effectively reflects tumor perfusion but is prone to artifacts from metal, blood, and air. DCE, based on T1-weighted contrast-enhanced imaging, is less artifact-prone but requires advanced post-processing software, limiting widespread use. ASL does not require gadolinium injection but has lower spatial resolution, which can limit clinical utility.

Additionally, susceptibility-weighted imaging (SWI) has been used to observe microbleeds and assess radiation-induced microvascular changes. Peters et al.47 found that children develop punctate SWI lesions more rapidly and earlier than adults. In a longitudinal study of pediatric patients treated for brain tumors with proton therapy, the cumulative incidence of radiation-induced cerebral microbleeds, as detected by SWI, increased progressively over time—reaching 43% at one year and 83% by five years post-therapy. The occurrence of cerebral microbleeds correlated significantly with higher radiation doses, greater irradiated brain volume, and younger age at treatment (p < 0.01).48 MRS is a non-anatomical imaging technique that quantifies metabolite concentrations within specific brain regions. MRS enables non-invasive detection of metabolic changes, allowing potential quantification of radiation-induced neurotoxicity. In the healthy brain, metabolites such as N-acetylaspartate and choline show distinct peaks.49 Among 242 patients who underwent both MRS and PET-computed tomography (CT) examinations, the diagnostic accuracy of MRS was 81.8%, significantly higher than that of PET-CT (42.9%).50 Changes in neuronal and glial cell populations alter intracranial metabolite concentrations.51 The N-acetylaspartate/creatine ratio is higher in regions of radiation necrosis than in tumor regions, while choline/N-acetylaspartate and choline/creatine ratios are higher in recurrent tumors. Reduced choline levels, along with potentially elevated lipid and lactate signals, suggest radiation necrosis.52 However, the long scan times required for accurate assessment limit the routine clinical use of MRS.

Deoxyhemoglobin is more paramagnetic than oxyhemoglobin, serving as a natural contrast agent. When vascular damage following radiotherapy causes imbalances between oxygen uptake and cerebral circulation, MRI sequences sensitive to magnetic field inhomogeneity can detect signal changes around cortical vessels. This is known as blood oxygenation level-dependent (BOLD) contrast. BOLD allows functional localization studies without contrast agents while providing high spatial resolution. Signal changes theoretically depend on intracranial blood oxygenation, blood flow, hematocrit, and tissue oxygen uptake, with blood flow being the primary determinant. BOLD functional MRI includes task-based and resting-state fMRI. Due to its ability to non-invasively measure hemodynamic changes, BOLD fMRI enables assessment of local neuronal and synaptic activity and, with high spatial and temporal resolution, has been widely used in neurological, psychiatric, and psychological research.53,54 This technique may also be applicable for detecting RIBI.

In cases of cerebral radiation necrosis, conventional MRI typically shows ring-enhancing lesions at the treatment site with surrounding edema, which are non-specific and may also occur in tumor progression.55 Diagnostic uncertainty for radiation necrosis based on conventional imaging alone can reach up to 15%.56 With the increasing use of immunotherapy, this uncertainty is further heightened, as pseudoprogression related to immune responses is often indistinguishable from true tumor progression using contrast-enhanced MRI alone.55 Therefore, additional imaging modalities are needed to supplement conventional methods for accurate evaluation. Based on the above content and previous studies, we summarized the general diagnostic and treatment workflow for MRI in RIBI (Fig. 2).

MRI diagnostic imaging workflow for RIBI.
Fig. 2  MRI diagnostic imaging workflow for RIBI.

(a) The workflow begins with clinical screening in patients with a history of radiotherapy and new neurological symptoms, excluding acute metabolic or vascular events. (b) Conventional structural MRI (T1WI, T2WI, FLAIR) identifies white matter lesions, necrosis, and atrophy, with FLAIR being particularly sensitive for detecting demyelination. (c) Diffusion imaging (DWI, ADC, MD) aids in differentiating hypercellular tumor recurrence from necrosis, while DTI (FA, AD, RD) assesses demyelination and axonal loss. (d) Perfusion imaging (PWI: CBV, CBF; ASL; DCE; DSC) evaluates hemodynamic characteristics, distinguishing low perfusion in necrosis from hyperperfusion in recurrence. (e) Metabolic and functional imaging (MRS for NAA, Cho, Lac, Lip; SWI for microbleeds; BOLD-fMRI) provides adjunctive information. If available, (f) AI/radiomics analysis offers quantitative imaging features to assist in differentiating necrosis from tumor recurrence. Finally, findings are (g) integrated with clinical history and (h) quantitative parameters to generate a diagnostic report, with follow-up recommended for monitoring dynamic changes when necessary. AD, axial diffusivity; AI, artificial intelligence; ASL, arterial spin labeling; BOLD-fMRI, blood oxygen level–dependent functional magnetic resonance imaging; CBF, cerebral blood flow; CBV, cerebral blood volume; DCE, dynamic contrast-enhanced (MRI); DSC, dynamic susceptibility contrast (MRI); DTI, diffusion tensor imaging; FA, fractional anisotropy; FLAIR, fluid-attenuated inversion recovery; MRS, magnetic resonance spectroscopy; NAA, N-acetylaspartate; PWI, perfusion-weighted imaging; RD, radial diffusivity; RIBI, radiation-induced brain injury; SWI, susceptibility-weighted imaging; T1WI, T1-weighted imaging.

Application of PET/CT and single photon emission computed tomography (SPECT) in the evaluation of RIBI

PET imaging can reveal the metabolic and inflammatory status within radiotherapy-treated regions and serves as a critical adjunct in the assessment of RIBI. PET/CT differentiates tissues based on varying uptake of radiolabeled glucose (fluorodeoxyglucose (FDG)). Tissues with higher glucose metabolism demonstrate increased FDG uptake, while necrotic tissues show reduced glucose metabolism and consequently decreased FDG uptake. Most tumor tissues have elevated glucose metabolism and thus higher FDG uptake. SPECT provides three-dimensional images of the target organ through multi-angle scanning. Various radiotracers are available for SPECT imaging; however, compared to MRI and PET/CT, SPECT has disadvantages including relatively lower spatial resolution, higher radiation dose, and slower scanning speed.57 Commonly used tracers include:

  • 18F-FDG, which reflects glucose metabolism but has limited discriminatory ability due to high background metabolic activity in brain tissue58;

  • 18F-DPA714, which targets the translocator protein and can identify microglial activation, represents a novel method for assessing inflammatory status. Translocator protein PET has been widely used in recent years to explore neuroinflammatory mechanisms in RIBI, providing imaging support for targeted therapies.59

However, the use of radiolabeled glucose has limitations. For example, some low-grade intracranial tumors have low FDG uptake, leading to false-negative results in clinical practice. Therefore, exploration of additional tracers is necessary to improve differentiation of RIBI.

Radiomics and artificial intelligence analysis

In recent years, radiomics combined with machine learning has emerged as a forefront approach for diagnosing RIBI. By extracting high-dimensional quantitative imaging features (such as intensity, texture, and shape), classification models are established to assist in distinguishing tumor recurrence from radiation necrosis. In 2023, Salari et al.60 constructed a random forest model based on T1+C and FLAIR sequence images, achieving over 90% classification accuracy in the validation cohort, providing technical support for noninvasive and automated diagnosis of RIBI. Moreover, deep learning models have shown promising prospects in predicting and dynamically evaluating RIBI.

The integration of multimodal imaging technology has allowed the assessment of RIBI to evolve from traditional structural evaluation toward comprehensive analysis encompassing function, metabolism, microstructure, and inflammatory status. Each imaging modality possesses distinct advantages, and their rational combined application facilitates earlier detection, improved differential diagnosis, and precise guidance for clinical intervention strategies.

Treatment of RIBI

RIBI currently lacks standardized treatment guidelines, with clinical management mainly consisting of symptomatic support and empirical interventions. With increasing understanding of its pathogenesis, therapeutic strategies have gradually expanded from traditional glucocorticoid-based approaches to multi-target comprehensive treatments addressing vascular leakage inhibition, neuroinflammation suppression, neural repair promotion, and gut-brain axis modulation. The following section reviews recent advances in RIBI treatment from the perspectives of pharmacotherapy, neuromodulation techniques, and systemic interventions (Table 2).61–67

Table 2

Treatment strategies for radiation-induced brain injury

TreatmentMechanismEvidence levelIndicationsAdvantagesLimitations/adverse effectsReferences
BevacizumabInhibits VEGF, reduces angiogenesis and vascular permeability, improves imaging enhancement and edemaRCT (Level I)Symptomatic radiation necrosis with edemaRapid symptom relief; reduces steroid dependenceHigh cost; potential hypertension and bleeding riskLevin et al., 201161
Corticosteroids (dexamethasone)Inhibit the release of inflammatory mediators, restore BBB integrity, alleviate edemaClinical observation (Level II-III)Acute phase of radiation-induced brain edema; symptomatic reliefRapid onset; widely accessibleLong-term use associated with significant adverse effects (osteoporosis, infections)Meixner et al., 202362
Hyperbaric oxygen (HBO)Provides high-pressure oxygen to promote neovascularization and repairSmall cohort/animal studies (Level III)Early-stage radiation-induced injury or refractory cases with edemaPotential prophylactic benefit; non-systemic toxicityLack of large-scale RCTs; resource-intensiveHajikarimloo et al., 202463
Mesenchymal stem cells (MSCs)Suppress microglial pyroptosis and inflammation, secrete neurotrophic factors, promote repairAnimal models (Level III)Experimental therapy for chronic radiation-induced brain injuryMulti-target regulation; improves cognition and tissue structureDifficulties with autologous transplantation; mechanisms remain unclear; no human trialsPan et al., 202564
Endothelial progenitor cells (EPCs)Participate in endothelial repair, re-endothelialization, and angiogenesisMouse/in vitro studies (Level III)Preclinical use for vascular damage after irradiationSpecific action; stabilizes BBBIsolation and purification challenges; low transplantation efficiencyGoksu et al., 202465
TSPO inhibitors (e.g., XBD173, PK11195)Target microglial activation, suppress neuroinflammation; dual role in diagnosis and therapyAnimal studies (Level III)Imaging and potential treatment of neuroinflammation in radiation-induced injurySmall molecules; easy delivery; allows imaging trackingLimited clinical validation; potential unknown adverse effectsZhang et al., 202366
Gut-brain axis interventions (probiotics/FMT)Modulate gut microbiota to reduce central inflammation and cognitive dysfunctionAnimal studies (Level III)Exploratory therapy for chronic cognitive impairment post-radiationNon-invasive; easily scalableComplex mechanisms; lacks human validationLuo et al., 202267

Pharmacological treatment

Bevacizumab is the only treatment demonstrated to be effective against cerebral radiation necrosis in randomized controlled trials.68 Preclinical studies have shown that intra-arterial administration of bevacizumab can successfully treat cerebral radiation necrosis, with effects lasting up to 8.5 months post-treatment. Pan et al.69 reported that bevacizumab relieved clinical symptoms in over 70% of RIBI patients, with MRI demonstrating improved perfusion and lesion shrinkage.

Corticosteroids are the first-line drugs for treating cerebral radiation necrosis. Steroids inhibit the release of cytokines and inflammatory mediators from necrotic tissue, reducing cytokine-induced inflammatory responses while improving BBB integrity and alleviating cerebral edema. Dexamethasone is commonly prescribed for RIBI. Short-term corticosteroid use can effectively reduce symptoms of cerebral radiation necrosis.70 However, clinicians should be alert to adverse effects associated with long-term use, and alternative treatment options need to be explored.

Sildenafil and simvastatin exhibit antioxidant and anti-inflammatory properties, acting respectively as nitric oxide donors and Tetrahydrobiopterin (BH4) regulators. Combined administration of sildenafil and simvastatin modulates the indoleamine 2,3-dioxygenase/kynurenine pathway through nitric oxide donor/BH4 regulatory activity, exerting anti-inflammatory and antioxidant effects, and provides neuroprotection against RIBI.71

Preclinical studies have explored the use of anticoagulants (heparin, warfarin),72 pentoxifylline,73 and metformin for RIBI management.74 These agents act through mechanisms including improving microcirculation, reducing fibrosis, promoting neuronal survival, and attenuating neuroinflammation. However, further studies are needed to confirm their safety and efficacy in clinical settings.

Hyperbaric oxygen therapy

Most case reports show improvement of radiation necrosis symptoms with hyperbaric oxygen (HBO) therapy, which also reduces corticosteroid usage.75 HBO effects have been observed to persist for extended periods76; however, randomized clinical trial data are lacking.

The basic principle of HBO in treating RIBI is that increased oxygen concentration stimulates angiogenesis, restoring blood supply to necrotic lesions and facilitating repair. Patients undergo treatment in a chamber at 2.5 atmospheres absolute, receiving 100% oxygen up to five times per week, with treatment cycles repeated up to 40 sessions. Studies indicate that prophylactic HBO after 20 sessions of SRS can reduce the incidence of brain radiation necrosis from 20% to 11% within one week post-treatment.77 Nonetheless, no prospective randomized controlled trials have definitively confirmed HBO’s therapeutic efficacy in RIBI.

Stem cell therapy

Preclinical studies indicate that mesenchymal stem cells contribute to vascular system repair in various organs and tissues. Strong evidence supports the role of endothelial progenitor cells in re-endothelialization, angiogenesis, and endothelial repair.78 Other studies report that transplanted bone marrow stem cells influence cytokine secretion,78 monocyte maturation and/or recruitment, and macrophage migration to injury sites,79 which may impede tissue repair. These findings warrant further investigation into the roles of endothelial progenitor cells and transplanted mesenchymal stem cells in vascular recovery.

Post-radiotherapy reductions in pericyte populations have been observed and may contribute to cognitive dysfunction. Pericytes are a heterogeneous group of mural cells associated with microvasculature and play critical roles in endothelial proliferation, BBB integrity, capillary constriction, and blood flow regulation.80,81 Mesenchymal stem cells differentiate into cell types constituting the neurovascular unit, including pericytes. Therefore, recruitment of bone marrow-derived pericytes to the brain following chronic systemic hypoxia requires further exploration. Extensive research is still needed to elucidate the reparative potential of mesenchymal stem cells in RIBI.

Biological therapy

Some researchers have identified astrocyte senescence and astrocyte-derived neuroinflammation as potential triggers of RIBI. For example, studies on Δ133p53 have shown that Δ133p53 inhibits full-length p53 and modulates factors such as p21 and IL-6,82–84 suppressing inflammatory processes. These inflammatory mediators have been implicated in radiation injury and neurotoxicity. Although the interactive mechanisms among these factors are not fully elucidated, findings suggest that preventing astrocyte senescence and inhibiting astrocyte-mediated neuroinflammation by inducing the p53 isoform Δ133p53 may hold therapeutic potential.82

Neuromodulation and rehabilitation interventions

Neuromodulation techniques are emerging as important non-pharmacological interventions for cognitive impairment and emotional disturbances in RIBI.83,84

  • Transcranial magnetic stimulation: Low-frequency repetitive transcranial magnetic stimulation has been applied in post-stroke cognitive rehabilitation, with preliminary studies showing improvements in attention and executive function in RIBI patients and demonstrating good safety profiles.85

  • Functional MRI neurofeedback: Training patients to regulate their own brain activity and enhance activation in specific brain regions has shown potential when combined with cognitive training and radiotherapy rehabilitation.86

Gut-brain axis-based intervention strategies

The gut-brain axis refers to the bidirectional communication network between the gut and the brain, serving as a key pathway linking the gut microbiota with neurological function.87

Recent studies have found that radiotherapy alters gut microbiota composition, influencing brain inflammation and neurological function, suggesting a potential role of the gut-brain axis in RIBI pathogenesis.88 In irradiated mouse models, gut dysbiosis is significant and accompanied by cognitive impairment and excessive microglial activation.89 Fecal microbiota transplantation and probiotic interventions partially reverse neural damage in animal experiments, indicating a promising therapeutic direction.90 Numerous studies have demonstrated a close association between the gut microbiota and various neurological disorders, including Alzheimer’s disease, traumatic brain injury, and Parkinson’s disease. Interventions targeting the gut microbiota have shown potential in altering the progression of these conditions. Therefore, the gut microbiota represents a promising therapeutic target for neurological diseases. In patients undergoing intracranial radiotherapy, disruption of the BBB increases the likelihood that gut microbiota and its metabolites may influence the nervous system. Although treatment strategies for RIBI are still under investigation, targeted modulation of the gut microbiota offers a novel therapeutic avenue.

Challenges and future research directions

Despite significant advances in the imaging diagnosis and treatment of RIBI in recent years, its clinical management still faces numerous challenges that require breakthroughs in the following areas:

Lack of unified diagnostic criteria and imaging specificity

Currently, the diagnosis of RIBI relies primarily on a combination of imaging findings and clinical experience, with no standardized diagnostic workflow or grading system established. Conventional MRI often has considerable limitations in detecting early lesion changes and differentiating them from tumor recurrence. Although multimodal imaging shows promise in improving diagnostic accuracy, its clinical application is hindered by complex image fusion processes, lack of unified technical standards, and high dependency on subjective interpretation.

Furthermore, data integration across different imaging modalities lacks a unified platform. In particular, the generalizability and clinical interpretability of radiomics and machine learning models in artificial intelligence-assisted diagnosis remain inadequate.

Limited therapeutic options and lack of mechanism-driven interventions

Current RIBI treatments mainly focus on glucocorticoids and symptomatic management, with few interventions targeting the underlying pathological mechanisms. Although bevacizumab can alleviate symptoms in the short term, its long-term efficacy and safety require validation in large-scale studies. Emerging strategies, including anti-inflammatory, antioxidant, neurotrophic, and gut-brain axis interventions, are mostly at the experimental stage, without an established, systematic evidence-based therapeutic framework.

Absence of early biomarkers and risk prediction models

The irreversible nature of RIBI underscores the importance of early identification and intervention. However, reliable predictive indicators and sensitive molecular imaging biomarkers are still lacking. Developing individualized risk prediction models by integrating clinical, imaging, blood-based, and genetic data represents a critical direction for future preventive intervention research.

Insufficient interdisciplinary collaborative research

RIBI involves multiple disciplines, including radiotherapy physics, neuroimaging, neurobiology, and clinical neuroscience. Current research is characterized by fragmented efforts with limited multi-center, large-sample, interdisciplinary collaboration, impeding the integrated advancement of mechanistic understanding, imaging techniques, and therapeutic interventions.

Future research directions

To address these shortcomings, future efforts should focus on: (1) Defining diagnostic standards: Establish consensus RIBI grading frameworks and integrate multimodal imaging data into interoperable workflows. (2) Targeting pathophysiology: Advance research on agents that modulate microglial activation or oxidative injury, such as RAS inhibitors that attenuate neuroinflammation in preclinical models.1 (3) Identifying predictive biomarkers: Validate blood and neuroimaging indicators of early RIBI risk (e.g., IL-1β, tumor necrosis factor-α, glial fibrillary acidic protein) and develop multivariable predictive models incorporating clinical, molecular, and imaging data. (4) Fostering multidisciplinary, multicenter research: Encourage collaborative networks that unify preclinical mechanistic insights, standardized imaging protocols, and therapeutic trials to translate findings into clinical practice.

Conclusions

RIBI remains a significant late complication of cranial radiotherapy, impacting cognition and quality of life. It is regarded as a complex and pressing complication in neuro-oncology, presenting persistent challenges in diagnosis and management. Multimodal imaging, including structural MRI, diffusion-based modalities, perfusion imaging, MRS/PET, and artificial intelligence-driven radiomics, has significantly enhanced early detection and differentiation of RIBI from tumor recurrence. However, most therapeutic strategies, including corticosteroids, bevacizumab, and experimental agents, remain largely palliative and lack high-level evidence for long-term benefit. Future efforts should focus on standardizing imaging workflows, developing validated predictive biomarkers, and conducting rigorous clinical trials of neuroprotective and neuromodulatory therapies to enable precision management of RIBI.

Declarations

Acknowledgement

None.

Funding

None.

Conflict of interest

The authors have no conflicts of interest to declare.

Authors’ contributions

Study concept and design (XC), manuscript writing (XW, SL), data analysis (XW, FY). All authors approved the manuscript prior to submission.

References

  1. Turnquist C, Harris BT, Harris CC. Radiation-induced brain injury: current concepts and therapeutic strategies targeting neuroinflammation. Neurooncol Adv 2020;2(1):vdaa057 View Article PubMed/NCBI
  2. Sreetharan S, Thome C, Tharmalingam S, Jones DE, Kulesza AV, Khaper N, et al. Ionizing Radiation Exposure During Pregnancy: Effects on Postnatal Development and Life. Radiat Res 2017;187(6):647-658 View Article PubMed/NCBI
  3. Qiu Y, Guo Z, Lin X, Li J, Li Z, Han L, et al. Standard radiotherapy for patients with nasopharyngeal carcinoma results in progressive tract-specific brain white matter alterations: A one-year follow-up via diffusion tensor imaging. Radiother Oncol 2021;159:255-264 View Article PubMed/NCBI
  4. Yang B, Ren BX, Tang FR. Prenatal irradiation-induced brain neuropathology and cognitive impairment. Brain Dev 2017;39(1):10-22 View Article PubMed/NCBI
  5. Demaria M, O’Leary MN, Chang J, Shao L, Liu S, Alimirah F, et al. Cellular Senescence Promotes Adverse Effects of Chemotherapy and Cancer Relapse. Cancer Discov 2017;7(2):165-176 View Article PubMed/NCBI
  6. Jiao Y, Cao F, Liu H. Radiation-induced Cell Death and Its Mechanisms. Health Phys 2022;123(5):376-386 View Article PubMed/NCBI
  7. Muñoz-Espín D, Serrano M. Cellular senescence: from physiology to pathology. Nat Rev Mol Cell Biol 2014;15(7):482-496 View Article PubMed/NCBI
  8. Wang Y, Boerma M, Zhou D. Ionizing Radiation-Induced Endothelial Cell Senescence and Cardiovascular Diseases. Radiat Res 2016;186(2):153-161 View Article PubMed/NCBI
  9. Turnquist C, Beck JA, Horikawa I, Obiorah IE, Von Muhlinen N, Vojtesek B, et al. Radiation-induced astrocyte senescence is rescued by Δ133p53. Neuro Oncol 2019;21(4):474-485 View Article PubMed/NCBI
  10. Li YQ, Chen P, Jain V, Reilly RM, Wong CS. Early radiation-induced endothelial cell loss and blood-spinal cord barrier breakdown in the rat spinal cord. Radiat Res 2004;161(2):143-152 View Article PubMed/NCBI
  11. Kiss T, Ungvari A, Gulej R, Nyúl-Tóth Á, Tarantini S, Benyo Z, et al. Whole brain irradiation-induced endothelial dysfunction in the mouse brain. Geroscience 2024;46(1):531-541 View Article PubMed/NCBI
  12. Zhou H, Liu Z, Liu J, Wang J, Zhou D, Zhao Z, et al. Fractionated radiation-induced acute encephalopathy in a young rat model: cognitive dysfunction and histologic findings. AJNR Am J Neuroradiol 2011;32(10):1795-1800 View Article PubMed/NCBI
  13. Tanikawa S, Kato Y, Tanino M, Terasaka S, Kurokawa Y, Arai N, et al. Autopsy report of a late delayed radiation injury after a period of 45 years. Neuropathology 2019;39(2):106-110 View Article PubMed/NCBI
  14. Zhang S, Li M, Qiu Y, Wu J, Xu X, Ma Q, et al. Enhanced VEGF secretion and blood-brain barrier disruption: Radiation-mediated inhibition of astrocyte autophagy via PI3K-AKT pathway activation. Glia 2024;72(3):568-587 View Article PubMed/NCBI
  15. Brown WR, Thore CR, Moody DM, Robbins ME, Wheeler KT. Vascular damage after fractionated whole-brain irradiation in rats. Radiat Res 2005;164(5):662-668 View Article PubMed/NCBI
  16. Xu M, Fan Q, Zhang J, Chen Y, Xu R, Chen L, et al. NFAT3/c4-mediated excitotoxicity in hippocampal apoptosis during radiation-induced brain injury. J Radiat Res 2017;58(6):827-833 View Article PubMed/NCBI
  17. Thabet NM, Rashed ER, Abdel-Rafei MK, Moustafa EM. Modulation of the Nitric Oxide/BH4 Pathway Protects Against Irradiation-Induced Neuronal Damage. Neurochem Res 2021;46(7):1641-1658 View Article PubMed/NCBI
  18. Lee WH, Sonntag WE, Mitschelen M, Yan H, Lee YW. Irradiation induces regionally specific alterations in pro-inflammatory environments in rat brain. Int J Radiat Biol 2010;86(2):132-144 View Article PubMed/NCBI
  19. Marmary Y, Adar R, Gaska S, Wygoda A, Maly A, Cohen J, et al. Radiation-Induced Loss of Salivary Gland Function Is Driven by Cellular Senescence and Prevented by IL6 Modulation. Cancer Res 2016;76(5):1170-1180 View Article PubMed/NCBI
  20. Matt S, Hofmann TG. The DNA damage-induced cell death response: a roadmap to kill cancer cells. Cell Mol Life Sci 2016;73(15):2829-2850 View Article PubMed/NCBI
  21. Chow BM, Li YQ, Wong CS. Radiation-induced apoptosis in the adult central nervous system is p53-dependent. Cell Death Differ 2000;7(8):712-720 View Article PubMed/NCBI
  22. Xiao M, Li X, Wang L, Lin B, Zhai M, Hull L, et al. Skin Wound following Irradiation Aggravates Radiation-Induced Brain Injury in a Mouse Model. Int J Mol Sci 2023;24(13):10701 View Article PubMed/NCBI
  23. Liu P, Fu M, Liu D, Chao T, Zhang J. Mechanisms of Radiation-induced Brain Injury in Mice Based on Bioinformatics Analysis. Radiat Res 2025;203(5):321-332 View Article PubMed/NCBI
  24. Wang TM, Shen GP, Chen MY, Zhang JB, Sun Y, He J, et al. Genome-Wide Association Study of Susceptibility Loci for Radiation-Induced Brain Injury. J Natl Cancer Inst 2019;111(6):620-628 View Article PubMed/NCBI
  25. Wen DW, Lin L, Mao YP, Chen CY, Chen FP, Wu CF, et al. Normal tissue complication probability (NTCP) models for predicting temporal lobe injury after intensity-modulated radiotherapy in nasopharyngeal carcinoma: A large registry-based retrospective study from China. Radiother Oncol 2021;157:99-105 View Article PubMed/NCBI
  26. Sun Y, Zhou GQ, Qi ZY, Zhang L, Huang SM, Liu LZ, et al. Radiation-induced temporal lobe injury after intensity modulated radiotherapy in nasopharyngeal carcinoma patients: a dose-volume-outcome analysis. BMC Cancer 2013;13:397 View Article PubMed/NCBI
  27. Iorio-Morin C, Mercure-Cyr R, Figueiredo G, Touchette CJ, Masson-Côté L, Mathieu D. Repeat stereotactic radiosurgery for the management of locally recurrent brain metastases. J Neurooncol 2019;145(3):551-559 View Article PubMed/NCBI
  28. Kim JW, Park HR, Lee JM, Kim JW, Chung HT, Kim DG, et al. Fractionated Stereotactic Gamma Knife Radiosurgery for Large Brain Metastases: A Retrospective, Single Center Study. PLoS One 2016;11(9):e0163304 View Article PubMed/NCBI
  29. Armstrong C, Ruffer J, Corn B, DeVries K, Mollman J. Biphasic patterns of memory deficits following moderate-dose partial-brain irradiation: neuropsychologic outcome and proposed mechanisms. J Clin Oncol 1995;13(9):2263-2271 View Article PubMed/NCBI
  30. Gonzalez J, Kumar AJ, Conrad CA, Levin VA. Effect of bevacizumab on radiation necrosis of the brain. Int J Radiat Oncol Biol Phys 2007;67(2):323-326 View Article PubMed/NCBI
  31. Lehrer EJ, Khosla AA, Ozair A, Gurewitz J, Bernstein K, Kondziolka D, et al. Immune checkpoint inhibition and single fraction stereotactic radiosurgery in brain metastases from non-small cell lung cancer: an international multicenter study of 395 patients. J Neurooncol 2023;165(1):63-77 View Article PubMed/NCBI
  32. Gan C, Li W, Xu J, Pang L, Tang L, Yu S, et al. Advances in the study of the molecular biological mechanisms of radiation-induced brain injury. Am J Cancer Res 2023;13(8):3275-3299 PubMed/NCBI
  33. Wu X, Ya J, Zhou D, Ding Y, Ji X, Meng R. Pathogeneses and Imaging Features of Cerebral White Matter Lesions of Vascular Origins. Aging Dis 2021;12(8):2031-2051 View Article PubMed/NCBI
  34. Masch WR, Wang PI, Chenevert TL, Junck L, Tsien C, Heth JA, et al. Comparison of Diffusion Tensor Imaging and Magnetic Resonance Perfusion Imaging in Differentiating Recurrent Brain Neoplasm From Radiation Necrosis. Acad Radiol 2016;23(5):569-576 View Article PubMed/NCBI
  35. Nael K, Bauer AH, Hormigo A, Lemole M, Germano IM, Puig J, et al. Multiparametric MRI for Differentiation of Radiation Necrosis From Recurrent Tumor in Patients With Treated Glioblastoma. AJR Am J Roentgenol 2018;210(1):18-23 View Article PubMed/NCBI
  36. Tringale KR, Nguyen TT, Karunamuni R, Seibert T, Huynh-Le MP, Connor M, et al. 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(4):773-783 View Article PubMed/NCBI
  37. Connor M, Karunamuni R, McDonald C, Seibert T, White N, Moiseenko V, et al. Regional susceptibility to dose-dependent white matter damage after brain radiotherapy. Radiother Oncol 2017;123(2):209-217 View Article PubMed/NCBI
  38. Connor M, Karunamuni R, McDonald C, White N, Pettersson N, Moiseenko V, et al. Dose-dependent white matter damage after brain radiotherapy. Radiother Oncol 2016;121(2):209-216 View Article PubMed/NCBI
  39. Chawla S, Wang S, Kim S, Sheriff S, Lee P, Rengan R, et al. Radiation injury to the normal brain measured by 3D-echo-planar spectroscopic imaging and diffusion tensor imaging: initial experience. J Neuroimaging 2015;25(1):97-104 View Article PubMed/NCBI
  40. Makola M, Douglas Ris M, Mahone EM, Yeates KO, Cecil KM. Long-term effects of radiation therapy on white matter of the corpus callosum: a diffusion tensor imaging study in children. Pediatr Radiol 2017;47(13):1809-1816 View Article PubMed/NCBI
  41. Chapman CH, Zhu T, Nazem-Zadeh M, Tao Y, Buchtel HA, Tsien CI, et al. Diffusion tensor imaging predicts cognitive function change following partial brain radiotherapy for low-grade and benign tumors. Radiother Oncol 2016;120(2):234-240 View Article PubMed/NCBI
  42. Hope TR, Vardal J, Bjørnerud A, Larsson C, Arnesen MR, Salo RA, et al. Serial diffusion tensor imaging for early detection of radiation-induced injuries to normal-appearing white matter in high-grade glioma patients. J Magn Reson Imaging 2015;41(2):414-423 View Article PubMed/NCBI
  43. Belka C, Budach W, Kortmann RD, Bamberg M. Radiation induced CNS toxicity—molecular and cellular mechanisms. Br J Cancer 2001;85(9):1233-1239 View Article PubMed/NCBI
  44. Nilsen LB, Digernes I, Grøvik E, Saxhaug C, Latysheva A, Geier O, et al. Responses in the diffusivity and vascular function of the irradiated normal brain are seen up until 18 months following SRS of brain metastases. Neurooncol Adv 2020;2(1):vdaa028 View Article PubMed/NCBI
  45. Lee MC, Cha S, Chang SM, Nelson SJ. Dynamic susceptibility contrast perfusion imaging of radiation effects in normal-appearing brain tissue: changes in the first-pass and recirculation phases. J Magn Reson Imaging 2005;21(6):683-693 View Article PubMed/NCBI
  46. Jajodia A, Goel V, Goyal J, Patnaik N, Khoda J, Pasricha S, et al. Combined Diagnostic Accuracy of Diffusion and Perfusion MR Imaging to Differentiate Radiation-Induced Necrosis from Recurrence in Glioblastoma. Diagnostics (Basel) 2022;12(3):718 View Article PubMed/NCBI
  47. Peters S, Pahl R, Claviez A, Jansen O. Detection of irreversible changes in susceptibility-weighted images after whole-brain irradiation of children. Neuroradiology 2013;55(7):853-859 View Article PubMed/NCBI
  48. Kralik SF, Mereniuk TR, Grignon L, Shih CS, Ho CY, Finke W, et al. Radiation-Induced Cerebral Microbleeds in Pediatric Patients With Brain Tumors Treated With Proton Radiation Therapy. Int J Radiat Oncol Biol Phys 2018;102(5):1465-1471 View Article PubMed/NCBI
  49. Wang Z, Zimmerman RA, Sauter R. Proton MR spectroscopy of the brain: clinically useful information obtained in assessing CNS diseases in children. AJR Am J Roentgenol 1996;167(1):191-199 View Article PubMed/NCBI
  50. Travers S, Joshi K, Miller DC, Singh A, Nada A, Biedermann G, et al. Reliability of Magnetic Resonance Spectroscopy and Positron Emission Tomography Computed Tomography in Differentiating Metastatic Brain Tumor Recurrence from Radiation Necrosis. World Neurosurg 2021;151:e1059-e1068 View Article PubMed/NCBI
  51. Lind A, Boraxbekk CJ, Petersen ET, Paulson OB, Siebner HR, Marsman A. Regional Myo-Inositol, Creatine, and Choline Levels Are Higher at Older Age and Scale Negatively with Visuospatial Working Memory: A Cross-Sectional Proton MR Spectroscopy Study at 7 Tesla on Normal Cognitive Ageing. J Neurosci 2020;40(42):8149-8159 View Article PubMed/NCBI
  52. Anbarloui MR, Ghodsi SM, Khoshnevisan A, Khadivi M, Abdollahzadeh S, Aoude A, et al. Accuracy of magnetic resonance spectroscopy in distinction between radiation necrosis and recurrence of brain tumors. Iran J Neurol 2015;14(1):29-34 PubMed/NCBI
  53. Greicius MD, Krasnow B, Reiss AL, Menon V. Functional connectivity in the resting brain: a network analysis of the default mode hypothesis. Proc Natl Acad Sci U S A 2003;100(1):253-258 View Article PubMed/NCBI
  54. Vanni S, Sharifian F, Heikkinen H, Vigário R. Modeling fMRI signals can provide insights into neural processing in the cerebral cortex. J Neurophysiol 2015;114(2):768-780 View Article PubMed/NCBI
  55. Mayo ZS, Halima A, Broughman JR, Smile TD, Tom MC, Murphy ES, et al. Radiation necrosis or tumor progression? A review of the radiographic modalities used in the diagnosis of cerebral radiation necrosis. J Neurooncol 2023;161(1):23-31 View Article PubMed/NCBI
  56. Siu A, Wind JJ, Iorgulescu JB, Chan TA, Yamada Y, Sherman JH. Radiation necrosis following treatment of high grade glioma—a review of the literature and current understanding. Acta Neurochir (Wien) 2012;154(2):191-201 View Article PubMed/NCBI
  57. Furuse M, Nonoguchi N, Yamada K, Shiga T, Combes JD, Ikeda N, et al. Radiological diagnosis of brain radiation necrosis after cranial irradiation for brain tumor: a systematic review. Radiat Oncol 2019;14(1):28 View Article PubMed/NCBI
  58. Brabazon F, Wilson CM, Shukla DK, Mathur S, Jaiswal S, Bermudez S, et al. FDG-PET Combined with MRI Elucidates the Pathophysiology of Traumatic Brain Injury in Rats. J Neurotrauma 2017;34(5):1074-1085 View Article PubMed/NCBI
  59. Wang Y, Yue X, Kiesewetter DO, Niu G, Teng G, Chen X. PET imaging of neuroinflammation in a rat traumatic brain injury model with radiolabeled TSPO ligand DPA-714. Eur J Nucl Med Mol Imaging 2014;41(7):1440-1449 View Article PubMed/NCBI
  60. Salari E, Elsamaloty H, Ray A, Hadziahmetovic M, Parsai EI. Differentiating Radiation Necrosis and Metastatic Progression in Brain Tumors Using Radiomics and Machine Learning. Am J Clin Oncol 2023;46(11):486-495 View Article PubMed/NCBI
  61. Levin VA, Bidaut L, Hou P, Kumar AJ, Wefel JS, Bekele BN, et al. Randomized double-blind placebo-controlled trial of bevacizumab therapy for radiation necrosis of the central nervous system. Int J Radiat Oncol Biol Phys 2011;79(5):1487-1495 View Article PubMed/NCBI
  62. Meixner E, Hörner-Rieber J, Lischalk JW, Eichkorn T, Krämer A, Sandrini E, et al. Management of initial and recurrent radiation-induced contrast enhancements following radiotherapy for brain metastases: Clinical and radiological impact of bevacizumab and corticosteroids. Clin Transl Radiat Oncol 2023;39:100600 View Article PubMed/NCBI
  63. Hajikarimloo B, Kavousi S, Jahromi GG, Mehmandoost M, Oraee-Yazdani S, Fahim F. Hyperbaric Oxygen Therapy as an Alternative Therapeutic Option for Radiation-Induced Necrosis Following Radiotherapy for Intracranial Pathologies. World Neurosurg 2024;186:51-61 View Article PubMed/NCBI
  64. Pan S, Wen Y, Liu Z, Xu K, Zhang N, Tong X, et al. Neuroprotective effects of bone marrow mesenchymal stem cells combined with mannitol on radiation-induced brain injury by regulating autophagy via the PI3K/AKT/mTOR signaling pathway. Brain Res Bull 2025;224:111326 View Article PubMed/NCBI
  65. Goksu AY, Kocanci FG, Akinci E, Demir-Dora D, Erendor F, Sanlioglu S, et al. Microglia cells treated with synthetic vasoactive intestinal peptide or transduced with LentiVIP protect neuronal cells against degeneration. Eur J Neurosci 2024;59(8):1993-2015 View Article PubMed/NCBI
  66. Zhang S, Deng Z, Qiu Y, Lu G, Wu J, Huang H. FGIN-1-27 Mitigates Radiation-induced Mitochondrial Hyperfunction and Cellular Hyperactivation in Cultured Astrocytes. Neuroscience 2023;535:23-35 View Article PubMed/NCBI
  67. Luo N, Zhu W, Li X, Fu M, Peng X, Yang F, et al. Impact of Gut Microbiota on Radiation-Associated Cognitive Dysfunction and Neuroinflammation in Mice. Radiat Res 2022;197(4):350-364 View Article PubMed/NCBI
  68. Voss M, Wenger KJ, Fokas E, Forster MT, Steinbach JP, Ronellenfitsch MW. Single-shot bevacizumab for cerebral radiation injury. BMC Neurol 2021;21(1):77 View Article PubMed/NCBI
  69. Pan D, Rong X, Chen D, Jiang J, Ng WT, Mai H, et al. Mortality of early treatment for radiation-induced brain necrosis in head and neck cancer survivors: A multicentre, retrospective, registry-based cohort study. EClinicalMedicine 2022;52:101618 View Article PubMed/NCBI
  70. Zielińska KA, Van Moortel L, Opdenakker G, De Bosscher K, Van den Steen PE. Endothelial Response to Glucocorticoids in Inflammatory Diseases. Front Immunol 2016;7:592 View Article PubMed/NCBI
  71. Feng Y, Feng Y, Gu L, Liu P, Cao J, Zhang S. The Critical Role of Tetrahydrobiopterin (BH4) Metabolism in Modulating Radiosensitivity: BH4/NOS Axis as an Angel or a Devil. Front Oncol 2021;11:720632 View Article PubMed/NCBI
  72. Glantz MJ, Burger PC, Friedman AH, Radtke RA, Massey EW, Schold SC. Treatment of radiation-induced nervous system injury with heparin and warfarin. Neurology 1994;44(11):2020-2027 View Article PubMed/NCBI
  73. Otluoglu GD, Yılmaz B, Ekinci G, Bayri Y, Bozkurt SU, Dağçınar A. Pentoxifylline and Vitamin E Can Restrict Radiation Necrosis via Vascular Pathways, Experimental Study in an Animal Model. World Neurosurg 2023;179:e530-e538 View Article PubMed/NCBI
  74. Xiang J, Lu Y, Quan C, Gao Y, Zhou G. Metformin Protects Radiation-Induced Early Brain Injury by Reducing Inflammation and DNA Damage. Brain Sci 2023;13(4):645 View Article PubMed/NCBI
  75. Mizrahi G, Wang Z, Leibovitz E. Hyperbaric Oxygen Treatment for Post Radiation Necrosis. Harefuah 2018;157(8):486-489 PubMed/NCBI
  76. Chung C, Bryant A, Brown PD. Interventions for the treatment of brain radionecrosis after radiotherapy or radiosurgery. Cochrane Database Syst Rev 2018;7(7):CD011492 View Article PubMed/NCBI
  77. Marx RE, Ehler WJ, Tayapongsak P, Pierce LW. Relationship of oxygen dose to angiogenesis induction in irradiated tissue. Am J Surg 1990;160(5):519-524 View Article PubMed/NCBI
  78. Yang M, Wei X, Li J, Heine LA, Rosenwasser R, Iacovitti L. Changes in host blood factors and brain glia accompanying the functional recovery after systemic administration of bone marrow stem cells in ischemic stroke rats. Cell Transplant 2010;19(9):1073-1084 View Article PubMed/NCBI
  79. Zhang Y, Ingram DA, Murphy MP, Saadatzadeh MR, Mead LE, Prater DN, et al. Release of proinflammatory mediators and expression of proinflammatory adhesion molecules by endothelial progenitor cells. Am J Physiol Heart Circ Physiol 2009;296(5):H1675-H1682 View Article PubMed/NCBI
  80. Luo N, Zhu W, Li X, Fu M, Zhang Y, Yang F, et al. Defective autophagy of pericytes enhances radiation-induced senescence promoting radiation brain injury. Neuro Oncol 2024;26(12):2288-2304 View Article PubMed/NCBI
  81. Galicich JH, French LA, Melby JC. Use of dexamethasone in treatment of cerebral edema associated with brain tumors. J Lancet 1961;81:46-53 PubMed/NCBI
  82. Fujita K, Mondal AM, Horikawa I, Nguyen GH, Kumamoto K, Sohn JJ, et al. p53 isoforms Delta133p53 and p53beta are endogenous regulators of replicative cellular senescence. Nat Cell Biol 2009;11(9):1135-1142 View Article PubMed/NCBI
  83. Turnquist C, Beck JA, Horikawa I, Obiorah IE, Von Muhlinen N, Vojtesek B, et al. Radiation-induced astrocyte senescence is rescued by Δ133p53. Neuro Oncol 2019;21(4):474-485 View Article PubMed/NCBI
  84. Turnquist C, Horikawa I, Foran E, Major EO, Vojtesek B, Lane DP, et al. p53 isoforms regulate astrocyte-mediated neuroprotection and neurodegeneration. Cell Death Differ 2016;23(9):1515-1528 View Article PubMed/NCBI
  85. Liu LY, Qin TZ, Guo L, Rong-Rong H, Jing YT, Lai PP, et al. The preventive and therapeutic effect of repetitive transcranial magnetic stimulation on radiation-induced brain injury in mice. Int J Radiat Biol 2022;98(8):1316-1329 View Article PubMed/NCBI
  86. He X, Moreno DR, Hou Z, Cheslack-Postava K, Jiang Y, Li T, et al. Connectivity based Real-Time fMRI Neurofeedback Training in Youth with a History of Major Depressive Disorder. ArXiv 2023 PubMed/NCBI
  87. Mayer EA, Nance K, Chen S. The Gut-Brain Axis. Annu Rev Med 2022;73:439-453 View Article PubMed/NCBI
  88. Palanivelu L, Chang CW, Li SJ, Liang YW, Lo YC, Chen YY. Interplay of Neuroinflammation and Gut Microbiota Dysbiosis in Alzheimer’s Disease Using Diffusion Kurtosis Imaging Biomarker in 3 × Tg-AD Mouse Models. ACS Chem Neurosci 2025;16(8):1511-1528 View Article PubMed/NCBI
  89. Zhao N, Chen QG, Chen X, Liu XT, Geng F, Zhu MM, et al. Intestinal dysbiosis mediates cognitive impairment via the intestine and brain NLRP3 inflammasome activation in chronic sleep deprivation. Brain Behav Immun 2023;108:98-117 View Article PubMed/NCBI
  90. Pan C, Zhang H, Zhang L, Chen L, Xu L, Xu N, et al. Surgery-induced gut microbial dysbiosis promotes cognitive impairment via regulation of intestinal function and the metabolite palmitic amide. Microbiome 2023;11(1):248 View Article PubMed/NCBI

About this Article

Cite this article
Wang X, Li S, Yu F, Cui X. State-of-the-art Multimodal Imaging and Therapeutic Strategies in Radiation-induced Brain Injury: A Comprehensive Review. Neurosurgical Subspecialties. Published online: Sep 18, 2025. doi: 10.14218/NSSS.2025.00028.
Copy        Export to RIS        Export to EndNote
Article History
Received Revised Accepted Published
May 30, 2025 August 9, 2025 August 13, 2025 September 18, 2025
DOI http://dx.doi.org/10.14218/NSSS.2025.00028
  • Neurosurgical Subspecialties
  • pISSN 0000-0000
  • eISSN 3067-6150
Back to Top

State-of-the-art Multimodal Imaging and Therapeutic Strategies in Radiation-induced Brain Injury: A Comprehensive Review

Xiaojie Wang, Shuang Li, Fangjing Yu, Xiaonan Cui
  • Reset Zoom
  • Download TIFF