Next Article in Journal
High-Voltage Overhead Power Line Fault Location Through Sequential Determination of Faulted Section
Previous Article in Journal
Benchmarking YOLOv8 to YOLOv11 Architectures for Real-Time Traffic Sign Recognition in Embedded 1:10 Scale Autonomous Vehicles
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Integrating Advanced Neuro-Oncology Imaging into Guideline-Directed Multimodal Therapy for Brain Metastases: Evaluating Comparative Treatment Effectiveness

1
The Legacy Heritage Cancer Center, Dr. Larry Norton Institute, Soroka Medical Center, Beer-Sheva 84105, Israel
2
Faculty of Health Sciences, Goldman Medical School, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
3
Neurosurgery Department, Soroka Medical Center, Beer-Sheva 84105, Israel
4
Neurosurgery Department, Hadassah Hebrew University Medical Center, Jerusalem 91200, Israel
5
Department of Oncology, The Emek Medical Centre, Afula 18341, Israel
6
Faculty of Health Sciences, Medical School for International Health, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
These authors also contributed equally to this work.
Technologies 2025, 13(11), 532; https://doi.org/10.3390/technologies13110532
Submission received: 17 October 2025 / Revised: 16 November 2025 / Accepted: 18 November 2025 / Published: 18 November 2025

Abstract

Background: Brain metastases (BM) are a common and serious complication in cancer patients, particularly those with lung, breast, or melanoma primaries. As systemic therapies extend survival, the incidence of BM has increased, necessitating improved diagnostic and treatment strategies. Recent advances in neuroimaging and therapy have significantly enhanced the ability to diagnose and manage these lesions with greater precision. Methods: This article summarizes current diagnostic imaging modalities—Magnetic Resonance Imaging (MRI), Positron Emission Tomography (PET), Perfusion-Weighted Imaging (PWI), and Magnetic Resonance Spectroscopy (MRS) and their roles in distinguishing tumor progression from treatment effects. It also compares the efficacy of therapeutic options including Whole-Brain Radiation Therapy (WBRT), Stereotactic Radiosurgery (SRS), and systemic therapies such as targeted drugs and immunotherapies. Outcomes were evaluated based on local tumor control and overall survival. Results: Advanced imaging techniques like PWI, MRS, and PET improve diagnostic accuracy by providing functional and metabolic information beyond standard MRI. Therapeutically, SRS offers better local control and fewer cognitive side effects than WBRT for patients with limited metastases. Targeted and immune-based therapies have shown improved survival in patients with specific genetic mutations, supporting a personalized treatment approach. Conclusions: The integration of advanced imaging and individualized therapies has improved diagnosis, treatment decisions, and outcomes in patients with brain metastases. Ongoing research is essential to refine these tools and approaches, further optimizing patient care and quality of life.

1. Introduction

Brain metastases (BM) represent the most common intracranial tumors in adults, posing a formidable challenge in oncology [1]. Historically associated with a dismal prognosis, the clinical landscape of BM is undergoing a profound transformation. Paradoxically, the incidence of BM is increasing, not as a result of more aggressive primary malignancies, but as a direct consequence of more effective systemic therapies. These treatments prolong patient survival, affording more time for cancer cells to seed the central nervous system (CNS), a sanctuary site often protected by the blood–brain barrier (BBB) [2,3]. This epidemiological shift has reframed BM from a pre-terminal event into a chronic, manageable condition for a growing number of patients. Consequently, the goals of care have evolved, moving beyond mere palliation to emphasize long-term intracranial disease control, preservation of neurocognitive function, and maintenance of quality of life [4].
The marked heterogeneity of BM—varying by primary tumor histology, molecular subtype, number, size, and location of lesions—precludes a “one-size-fits-all” therapeutic approach [1]. The central clinical challenge lies in selecting the optimal combination and sequence of local therapies (surgical resection, Stereotactic Radiosurgery, Whole-Brain Radiation Therapy) and systemic therapies (conventional chemotherapy, targeted agents, immunotherapy) for each individual patient [5,6]. This decision-making process is complex and requires a sophisticated diagnostic toolkit capable of not only detecting lesions but also characterizing their underlying biology and response to treatment. Conventional contrast-enhanced Magnetic Resonance Imaging (MRI), while the standard for detection, often falls short in the critical post-treatment setting, where distinguishing true tumor progression from treatment-induced effects like radiation necrosis is a frequent and consequential dilemma [7].
The objective of this article is to provide clinicians—including neuro-oncologists, medical oncologists, radiation oncologists, and neurosurgeons—with a comprehensive, evidence-based framework for integrating advanced neuro-imaging into the multidisciplinary management of BM. This article synthesizes the current information and literature on advanced imaging modalities and comparative adjuvant therapies, anchoring the discussion in the latest clinical practice guidelines from major international oncology organizations. By systematically connecting diagnostic findings to therapeutic strategies, this article aims to serve as a definitive resource for understanding and applying modern, personalized approaches to improve outcomes for patients with brain metastases.

2. Material and Methods

To ensure a comprehensive and current review, a literature search was conducted using the PubMed/MEDLINE and Embase databases for relevant articles published up to August 2025. The search strategy employed a combination of medical subject headings (MeSH) and keywords, including “brain metastases,” “neuroimaging,” “advanced magnetic resonance imaging,” “perfusion-weighted imaging,” “magnetic resonance spectroscopy,” “positron emission tomography,” “stereotactic radiosurgery,” “whole-brain radiation therapy,” “targeted therapy,” and “immunotherapy.” These terms were combined with keywords for specific primary cancers known to frequently metastasize to the brain, such as “non-small cell lung cancer,” “breast cancer,” and “melanoma.” The search was limited to studies published in English. Additionally, the clinical practice guidelines and relevant publications from major oncology societies were systematically reviewed and incorporated, including those from the National Comprehensive Cancer Network (NCCN), the American Society of Clinical Oncology (ASCO), the Society for Neuro-Oncology (SNO), the American Society for Radiation Oncology (ASTRO), and the European Society for Medical Oncology (ESMO) in partnership with the European Association of Neuro-Oncology (EANO).

3. The Multidisciplinary Treatment Landscape for Brain Metastases

The management of brain metastases requires a coordinated, multidisciplinary approach that integrates various therapeutic modalities. The selection and sequencing of these treatments are guided by patient-specific factors, including performance status, systemic disease control, and the number, size, and location of intracranial lesions, as well as the primary tumor’s molecular characteristics.

3.1. The Foundational Role of Local Control: Surgical Resection and Stereotactic Radiosurgery (SRS)

Effective local control of intracranial lesions is paramount to preventing neurological decline and improving survival. Surgery and SRS are the cornerstones of local therapy for limited brain metastases [8,9].

3.1.1. Surgical Resection

Surgical resection remains a primary treatment modality, particularly for patients with a single, accessible brain metastasis, especially when the lesion is large and causing significant mass effect or neurological symptoms [10]. According to consensus guidelines from ASCO and ESMO, surgery is strongly recommended in patients with good performance status and controlled or limited systemic disease [6]. It also serves a crucial diagnostic purpose when the primary cancer is unknown or if there is a suspicion of a non-metastatic lesion [6]. Furthermore, resection can be considered for radioresistant tumors, such as renal cell carcinoma or melanoma, where radiation alone may be less effective [10]. While a technical debate exists between en-bloc (one-piece) and piecemeal resection, some evidence suggests that en-bloc resection may reduce the risk of leptomeningeal dissemination, although it is not always technically feasible [11].

3.1.2. Stereotactic Radiosurgery (SRS)

SRS is an advanced radiation technique that delivers a highly conformal, high-dose ablative radiation dose to one or more metastatic lesions while minimizing exposure to the surrounding healthy brain tissue [10,11,12]. It has become a standard of care and a primary local therapy option, particularly for patients with a limited number of inoperable or smaller metastases (typically <3–4 cm in diameter) [12,13]. The principal advantage of SRS over WBRT is the significant reduction in neurotoxicity, leading to better preservation of long-term neurocognitive function—a critical consideration for patients with longer life expectancies [11,12,13].
The roles of surgery and SRS are not mutually exclusive; rather, they are increasingly intertwined in a synergistic fashion. Historically, WBRT was the standard adjuvant treatment following surgical resection to address microscopic residual disease and reduce the risk of recurrence. However, given the neurocognitive sequelae of WBRT, postoperative SRS delivered to the surgical cavity has emerged as the preferred strategy to improve local control [12,13]. This approach provides focused radiation to the area at highest risk of recurrence while sparing the rest of the brain. More recently, preoperative SRS has been explored as a novel strategy. Delivering SRS to an intact metastasis before resection may reduce the risk of intraoperative tumor cell spillage and subsequent leptomeningeal recurrence, a known complication of surgery [12,13]. This evolving paradigm demonstrates that the clinical decision is often not a simple choice of “surgery OR SRS,” but rather a carefully planned sequence of “surgery THEN SRS” or “SRS THEN surgery,” necessitating close collaboration between neurosurgeons and radiation oncologists from the outset to choreograph the optimal local treatment plan.

3.2. Whole-Brain Radiation Therapy (WBRT): Evolving Indications and Mitigation of Neurotoxicity

WBRT was historically the standard of care for patients with brain metastases, especially those with multiple lesions [14,15,16]. It treats both visible and microscopic disease throughout the entire brain. However, its use has declined substantially in recent years due to its strong association with significant, progressive, and often irreversible long-term neurocognitive decline, including memory loss and executive dysfunction [12,13,14,15,16]. As systemic therapies improve and patients live longer after a BM diagnosis, preserving quality of life and cognitive function has become a primary goal of treatment, leading to a re-evaluation of WBRT’s role.
Currently, guideline-informed practice reserves WBRT for specific clinical scenarios. It is most often considered for patients with numerous metastases (e.g., >10–15) that are not amenable to SRS, or as a palliative treatment for symptomatic patients with a poor performance status and limited life expectancy [12,13,14,15,16]. When WBRT is deemed necessary, clinical guidelines strongly recommend the implementation of strategies to mitigate its cognitive toxicity. These include hippocampal-avoidance WBRT (HA-WBRT), a technique that selectively spares the hippocampi—brain structures critical for memory formation—from high-dose radiation. Additionally, the administration of the drug memantine, an N-methyl-D-aspartate (NMDA) receptor antagonist, has been shown to delay cognitive decline in patients receiving WBRT (Table 1). These neuroprotective strategies are recommended for patients with an expected survival of at least four months [14,15,16].

3.3. The Paradigm Shift with Systemic Therapies

The efficacy of systemic therapies for brain metastases has historically been limited by the BBB, a specialized endothelial barrier that restricts the passage of many therapeutic agents, including most traditional cytotoxic chemotherapies, into the CNS [17,18,19,20,21,22]. Consequently, clinical practice guidelines generally do not recommend the routine use of cytotoxic chemotherapy, either alone or in combination with WBRT or SRS, for the management of brain metastases from most solid tumors, with only a few specific exceptions [17,18,19,20,21,22].
However, the modern era of oncology has ushered in a new class of CNS-penetrant agents that have revolutionized the management of BM [19,20,21,22]. The development of small-molecule tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors (ICIs) has fundamentally altered the treatment paradigm. These agents can effectively cross the BBB and achieve significant and durable intracranial responses. This has led to a paradigm shift where, for select patients—particularly those who are asymptomatic and have tumors with known targetable driver mutations (e.g., EGFR-mutant NSCLC) or high immunogenicity (e.g., melanoma)—upfront systemic therapy can be considered as the initial treatment. This approach can delay or, in some cases, even obviate the need for brain-directed local therapy, thereby avoiding the potential toxicities associated with radiation or surgery [19,20,21,22,23,24].

4. Foundational Imaging Modalities in Neuro-Oncology

Accurate diagnosis, precise delineation, and effective monitoring of brain tumors and metastases are critically dependent on advanced imaging modalities. These techniques provide distinct yet complementary information, ranging from anatomical detail to metabolic and physiological insights.

4.1. Magnetic Resonance Imaging (MRI): Principles and Diagnostic Utility

Magnetic Resonance Imaging (MRI) stands as a cornerstone in neuro-oncology diagnostics due to its unparalleled ability to visualize the intricate structures of the brain and spinal cord. Unlike X-rays, MRI operates by employing powerful magnetic fields, radio waves, and sophisticated computer processing, fundamentally distinguishing itself by not utilizing ionizing radiation. This absence of radiation exposure is a crucial advantage, making MRI the preferred imaging modality for patients requiring frequent monitoring for diagnosis or treatment evaluation, particularly within the brain. The non-ionizing nature of MRI allows for more aggressive and frequent surveillance strategies without adding to a patient’s cumulative radiation burden, thereby enabling earlier detection of recurrence or treatment-related changes and facilitating more personalized, long-term management [25].
To enhance image clarity and diagnostic precision, a special dye, typically a gadolinium-based contrast medium, is administered intravenously. This contrast agent alters the magnetic properties of nearby water molecules, causing areas of interest such as tumors, inflammation, infection, and specific blood vessels to appear brighter and more distinct on the images. This enhancement is particularly valuable for identifying the presence, location, size, and other characteristics of brain tumors, which are vital considerations for treatment planning [26].

4.1.1. Several Specialized MRI Techniques Are Employed for Comprehensive Brain Tumor Diagnosis and Monitoring

Intravenous (IV) Gadolinium-Enhanced MRI
This is a standard procedure where gadolinium accumulates around cancerous cells, making them more conspicuous for comparison with pre-contrast images [26].
Diffusion-Weighted Imaging (DWI)
This technique provides critical information about white matter tracts and helps evaluate the cellularity, nature, and structural integrity of brain tumors. It is also valuable in assessing conditions like stroke [26].
Perfusion Imaging (PWI)
This technique focuses on the microscopic blood vessels supplying a tumor, offering insights into its vascularity [26].
Functional MRI (fMRI)
During fMRI, patients perform specific tasks, which induce subtle blood flow changes in active brain regions. These changes are then visualized, providing crucial information about the location of areas responsible for critical functions such as speech and motor skills, aiding in surgical planning [26].
Magnetic Resonance Spectroscopy (MRS)
This advanced technique provides detailed information on the chemical composition of brain tissue. It helps in identifying cancerous cells and monitoring tumor progression by analyzing metabolite concentrations [26].
MRI is foundational in neuro-oncology, playing a pivotal role in accurately identifying the presence, location, size, and specific characteristics of brain tumors. It is indispensable for guiding treatment planning, monitoring the tumor’s response to therapy, and long-term surveillance. During the scan, patients lie still and may hear loud knocking and clicking sounds, for which earplugs or headphones are provided. Two-way communication with the technologist is maintained via an intercom system. While generally painless, the enclosed nature of some MRI machines can be challenging for claustrophobic patients, sometimes necessitating light or heavy sedation. The presence of metallic implants or objects on the body is a contraindication due to the strong magnetic fields generated during MRI examinations, and therefore a wide range of implants must be carefully assessed for MRI compatibility and safety. Many are considered safe and are verified through the MRI safety screening process prior to scanning, including abdominal aortic aneurysm stents, stapes or other middle-ear implants, implanted drug infusion pumps, neurostimulators or bone growth stimulators, surgical clips, wire sutures, screws, mesh materials, ocular or penile prostheses, joint replacements or other orthopedic prostheses, as well as various other internal mechanical devices. Notably, titanium—commonly used in many medical implants—is a paramagnetic material and is generally not affected by the magnetic field during MRI examinations [26,27,28].

4.2. Positron Emission Tomography (PET) Scan: Principles and Metabolic Insights

Positron Emission Tomography (PET) scanning is an advanced imaging technique widely employed in oncology, providing unique insights into metabolic activity, blood flow, and chemical composition within the body. This technique involves the administration of radiotracers, which can be injected, swallowed, or inhaled, and are absorbed by tissues based on their specific affinity. These tracers contain unstable nuclei that emit positrons, which then interact with electrons to produce gamma rays. A ring of detectors in the scanner captures these gamma rays, and a computer processes the data to generate a detailed 3D image of the tracer’s distribution. Areas of heightened metabolic activity exhibit increased tracer uptake, appearing as brighter spots on the scan [28,29,30].

4.2.1. Key Radiotracers Utilized in Neuro-Oncology

18F-Fluorodeoxyglucose (18F-FDG)
This is the most commonly used PET tracer, acting as a glucose analog absorbed by cells for metabolism. Cancerous tissues typically exhibit high metabolic activity due to the “Warburg effect”—increased glucose uptake and glycolysis even in the presence of oxygen—leading to significant absorption of 18F-FDG and bright spots on the scan. This characteristic makes 18F-FDG PET highly effective for diagnosing, staging, and monitoring various cancers, including the detection of metastases. However, its utility in brain imaging can be limited by high physiological uptake in normal gray matter and non-specific uptake in inflammatory lesions [31,32,33].
Amino Acid PET Tracers (e.g., [18F]FET, [11C]MET, [18F]FDOPA)
These tracers are particularly valuable in neuro-oncology due to their high tumor-to-brain contrast. This specificity arises from the increased expression of large neutral amino acid transporters (LAT1, LAT2) in gliomas and brain metastases, leading to relatively high uptake in neoplastic tissue and low uptake in healthy brain tissue. This metabolic specificity often makes them superior to 18F-FDG for certain brain tumor applications. While 18F-FDG is effective for systemic staging and identifying aggressive extracranial disease, amino acid tracers offer superior specificity within the complex brain environment by targeting unique amino acid transport mechanisms upregulated in tumors, enabling more precise delineation and differentiation. This guides the strategic selection of PET tracers based on the specific clinical question, ensuring that for nuanced intracranial questions like tumor boundary definition or recurrence versus necrosis, amino acid PET provides indispensable, higher-resolution metabolic information [33,34,35].

4.2.2. Clinical Applications of PET Scans in Neuro-Oncology Are Diverse and Critical

Diagnosis and Staging
PET scans can detect and stage most cancers, often before they are evident through other imaging modalities like CT or MRI. They are crucial for diagnosing aggressive tumors and tracking their spread [32].
Treatment Planning
Amino acid PET tracers are invaluable for precisely delineating tumor spread, which is essential for planning stereotactic biopsies, surgical resection, and radiotherapy. The metabolically active tumor burden, as assessed by amino acid PET, frequently extends beyond the volume of contrast enhancement seen on MRI, providing critical information for comprehensive treatment planning [32,33,34].
Monitoring Treatment Response
PET scans are utilized to assess the effectiveness of various treatments, including surgery, radiation therapy, and chemotherapy, by monitoring changes in metabolic activity over time. This helps physicians determine whether a tumor is responding to therapy. They can also identify “pseudoresponse” following antiangiogenic therapies, where MRI might show decreased enhancement without true clinical benefit [32].
Differentiation of Tumor Progression from Treatment-Related Changes
This is a critical application. Amino acid PET, particularly using [18F]FET or [18F]FDOPA, provides valuable diagnostic information for differentiating radiation injury from tumor relapse in both glioma and brain metastasis patients, with high diagnostic accuracy (80–90%). While [11C]MET has a slightly lower accuracy (around 75%) due to its higher affinity for inflammation, these tracers are also helpful in distinguishing true relapse from immunotherapy-induced inflammation, which can mimic progression on MRI [32,33,34].

4.3. Perfusion-Weighted Imaging (PWI): Assessing Tumor Vascularity

Perfusion-Weighted Imaging (PWI) is a specialized Magnetic Resonance Imaging (MRI) technique that provides crucial quantitative and qualitative information about cerebral blood flow (CBF) and tissue perfusion. This technique fundamentally relies on measuring signal changes within the brain tissue following the introduction of a contrast agent or by utilizing endogenous signals [35].

4.3.1. The Primary PWI Techniques Include

Dynamic Susceptibility Contrast (DSC) MRI
This is a widely used PWI technique involving the rapid intravenous injection of a paramagnetic contrast agent, typically a gadolinium-based compound. As the contrast agent passes through the brain’s microvasculature, it causes a temporary reduction in MR signal intensity. By rapidly acquiring a series of images during this “first pass,” clinicians can assess cerebral perfusion. The signal changes are then used to calculate hemodynamic parameters such as cerebral blood volume (CBV) and cerebral blood flow (CBF). Relative cerebral blood volume (rCBV) is often employed due to the inherent difficulties in absolute quantification [36].
Arterial Spin Labeling (ASL)
This method offers a non-invasive alternative as it uses magnetically labeled arterial blood as an endogenous contrast agent. ASL provides a quantitative measure of CBF without the need for exogenous contrast agents, eliminating concerns related to gadolinium administration [36].
PWI is instrumental in the diagnosis, characterization, and management of brain tumors by providing insights into their vascularity, grade, and potential response to treatment [37]. This technique offers a unique and complementary layer of information that goes beyond merely assessing BBB integrity, which is the primary focus of conventional contrast-enhanced MRI. While BBB disruption is a non-specific sign, PWI provides a functional assessment of the lesion’s microvascular environment, which is a direct indicator of tumor angiogenesis and metabolic demand. This distinction is crucial because it allows clinicians to differentiate between lesions that may appear similar on conventional imaging but have fundamentally different underlying pathologies, such as highly vascular but non-enhancing tumors, or treatment-related changes with BBB disruption but no increased neovascularity [38]. PWI thus enhances diagnostic precision, guides biopsy targeting to the most aggressive areas, and helps in monitoring treatment efficacy more accurately, particularly in scenarios where conventional imaging is ambiguous [36].

4.3.2. Clinical Utility of PWI in Neuro-Oncology Includes

Differentiation Between Tumor Types
Distinct perfusion patterns are observed across different brain tumor types [36].
High-Grade Gliomas
These typically exhibit increased CBV and CBF, reflecting their high vascularity and aggressive nature [36].
Low-Grade Gliomas and Benign Tumors
These generally show lower perfusion characteristics. However, it is important to note that some low-grade gliomas, such as pilocytic astrocytomas and oligodendrogliomas, can be inherently hypervascular, leading to elevated rCBV values despite their lower histological grade [36].
Lymphomas
These often present with lower CBV compared to high-grade gliomas. Lymphomas consistently show low rCBV despite often exhibiting strong homogeneous enhancement on conventional MRI, providing a key differentiating feature [36].
Hemangioblastomas
These tumors consistently demonstrate the highest rCBV ratios among brain tumors, significantly higher than other types, making PWI useful in differentiating them from other cystic lesions [36].
Metastases
The rCBV ratios of metastases are often similar to those of high-grade gliomas, typically showing high rCBV [36].
Assessment of Tumor Grade and Aggressiveness
Tumor perfusion characteristics are directly correlated with tumor grade and aggressiveness. Studies have consistently shown that higher CBV values are associated with higher tumor grades and a poorer prognosis. For instance, a statistically significant difference in rCBV ratios (p = 0.001) has been observed between high-grade gliomas (mean ± SD, 9.33 ± 3.96) and low-grade gliomas (mean ± SD, 3.64 ± 1.50) [39].
Identification of Tumor Boundaries and Infiltration
PWI is valuable in identifying the full extent of tumor infiltration into surrounding brain tissue. This information is critical for meticulous surgical planning and ensuring maximal, safe tumor resection [11].
Differentiation from Treatment-Related Changes
PWI is increasingly utilized to distinguish between true tumor progression and pseudo-progression or radiation necrosis. Meta-analyses have shown that the average rCBV in a contrast-enhancing lesion is significantly higher in tumor recurrence compared with radiation injury (pooled difference in means 2.18, 95% CI = 0.85 to 3.50, p = 0.001). The mean maximum rCBV was also significantly higher in the metastasis group compared to the radiation necrosis group (p = 0.048). This distinction is based on the principle that active tumor recurrence typically exhibits increased neovascularity and blood flow, whereas radiation necrosis is often characterized by hypovascularity or avascularity (Table 2) [15].

4.4. Magnetic Resonance Spectroscopy (MRS): Biochemical Characterization of Brain Lesions

Magnetic Resonance Spectroscopy (MRS) is a non-invasive diagnostic technique that measures biochemical changes within the brain, providing a metabolic fingerprint of tissue. Unlike conventional MRI, which provides anatomical images, MRS compares the chemical composition of normal brain tissue with that of abnormal or suspected tumor tissue. Performed on the same MRI machine, it involves a series of additional tests designed to measure the chemical metabolism within a lesion. MRS primarily analyzes molecules such as hydrogen ions or protons, with proton spectroscopy being the most commonly used method. This capability allows MRS to function as a non-invasive “biochemical biopsy,” providing a quantitative, differential diagnosis based on the underlying biochemical processes without the risks and costs associated with surgical biopsy. This is a profound advancement, enabling clinicians to distinguish between active cellular proliferation (characteristic of tumor recurrence) and tissue necrosis (characteristic of radiation injury), which has significant implications for patient management [28,40].
MRS quantifies the concentration of various metabolites, which are products of cellular metabolism, to differentiate between tissue types and pathological conditions. The frequency of these metabolites is measured in parts per million (ppm) and plotted as peaks on a graph [28].

4.4.1. Key Metabolites and Their Significance Include

N-acetyl aspartate (NAA): Primarily found in neurons, considered a marker of neuronal viability and density. Decreased NAA levels often indicate neuronal damage or loss [41].
Choline (Cho): A component of cell membranes, elevated Cho levels typically indicate increased cell membrane turnover, a hallmark of cellular proliferation and tumor aggressiveness [41].
Creatine (Cr): Involved in energy metabolism, often used as an internal reference standard due to its relatively stable concentration in healthy brain tissue [41].
Lactate: An indicator of anaerobic glycolysis, often elevated in highly aggressive tumors due to hypoxia [41].
Lipid: Indicates necrosis or cellular breakdown, often seen in high-grade tumors with central necrosis [41].
Amino acids, Myoinositol, Alanine: Other metabolites that can provide specific diagnostic clues [41].

4.4.2. Clinical Applications of MRS in Brain Tumor Diagnosis, Characterization, and Differentiation Are Crucial

Tumor Type and Aggressiveness
By analyzing metabolite ratios, MRS can help differentiate between various tumor types and assess their aggressiveness.
Glioma: Characteristically shows lower than normal NAA levels, elevated choline and lipid levels, and often the presence of lactate peaks [28].
Meningioma: May be indicated by elevated alanine levels [28,41].
Differentiation of Tumor Recurrence from Radiation Necrosis
This is one of the most significant applications of MRS. Conventional imaging often struggles to distinguish between these two outcomes, which have vastly different management implications.
Key Differentiating Feature: Radiation necrosis typically does not exhibit elevated choline levels, a stark contrast to active tumor recurrence where choline is usually elevated due to increased cell proliferation [41].

4.4.3. Diagnostic Accuracy (Meta-Analysis Findings)

MRS is effective in distinguishing recurrent brain tumors from necrosis. Meta-analyses have revealed that Cho/NAA, Cho/Cr, and NAA/Cr ratios are significantly better predictors of detected recurrent tumor. Specifically, an elevated choline/NAA ratio has high sensitivity and specificity for actual disease progression. A pooled difference in means for Cho/Cr ratio was significantly higher in tumor recurrence than in tumor necrosis (0.77, 95%CI = 0.57 to 0.98, p = 0.0001). Similarly, a significant difference was found in Cho/NAA ratios between recurrent tumor and necrosis (1.02, 95%CI = 0.03 to 2.00, p = 0.044). Among various imaging modalities studied for this purpose, MRS demonstrated the highest pooled sensitivity (90.7%), (Table 3) [40].
Patients preparing for an MRS test are advised to avoid caffeinated beverages and remove all metallic items. The procedure involves lying still on a movable bed, with a specialized “coil” placed over the area of interest. Patients will hear thumping sounds during image acquisition. Contrast dye (gadolinium) may be injected. The test can take slightly longer than a conventional MRI. MRI and MRS are generally very safe, with no known health risks associated with the magnetic field or radio waves. Allergic reactions to contrast agents are possible but rare. Metallic implants are contraindications, and pregnancy requires careful consideration [28].
Regarding the quantitative comparisons of sensitivity, specificity, and Area Under the Curve (AUC) for MRI, PWI, MRS, and PET in BM diagnosis are challenging due to significant variations in study methodologies, patient populations, and the specific radiotracers used. Available data suggest all are valuable diagnostic tools, with advanced techniques generally offering improved accuracy over conventional MRI alone, particularly in differentiating tumor recurrence from treatment effects, (Table 4), [42,43,44,45].

5. Advanced Imaging in the Critical Post-Treatment Setting: Differentiating Recurrence from Necrosis

One of the most frequent and challenging clinical dilemmas in neuro-oncology is the interpretation of new or enlarging contrast-enhancing lesions on follow-up MRI after local therapy, particularly SRS. These imaging changes can represent either true tumor progression, which requires prompt and aggressive intervention, or treatment-related effects such as radiation necrosis, a form of sterile inflammation and tissue death that may be managed conservatively. Conventional MRI, which primarily visualizes anatomy and BBB disruption, is often ambiguous in this setting, as both pathologies can appear strikingly similar [37]. Advanced imaging techniques that provide physiological and metabolic information beyond anatomy are indispensable for resolving this diagnostic uncertainty.

5.1. Perfusion-Weighted Imaging (PWI): Quantifying Neovascularity to Unmask True Progression

PWI is an advanced MRI technique that assesses tissue hemodynamics, providing insights into the microvascular environment of a lesion. The core principle underlying its utility in this context is that recurrent tumors, to sustain their growth, must develop new, often leaky and disorganized, blood vessels—a process known as neovascularization. This leads to a measurable increase in regional cerebral blood volume (CBV) and cerebral blood flow (CBF). In stark contrast, radiation necrosis is a pathological process characterized by endothelial damage, vascular occlusion, and tissue death, which typically results in hypoperfusion, or decreased CBV and CBF [38]. By quantifying these hemodynamic parameters, PWI can help differentiate metabolically active, highly vascularized recurrent tumors from avascular necrotic tissue. The clinical utility of this technique is supported by robust evidence. A key meta-analysis by Chuang et al. which analyzed data from ten studies, found that the relative cerebral blood volume (rCBV) in enhancing lesions was significantly higher in cases of tumor recurrence compared to radiation injury, with a pooled difference in means of 2.18 (95% CI = 0.85 to 3.50, p = 0.001) [39,41,46]. This quantitative evidence underscores PWI’s role in unmasking the neovascular signature of true tumor progression.

5.2. Magnetic Resonance Spectroscopy (MRS): A Non-Invasive Metabolic Biopsy

MRS is a non-invasive technique that provides a biochemical “fingerprint” of brain tissue by measuring the concentration of various metabolites. This metabolic profile offers a window into the underlying cellular processes within a lesion. The differentiation between recurrence and necrosis is based on distinct metabolic signatures. Recurrent tumors are characterized by high rates of cellular proliferation and membrane synthesis, which leads to a significantly elevated peak of Choline (Cho), a cell membrane component. Conversely, N-acetyl aspartate (NAA), a marker of neuronal viability, decreases in both tumor and necrotic tissue due to the destruction of normal neurons. Creatine (Cr), involved in cellular energy metabolism, remains relatively stable and is often used as an internal reference standard [30,31,32]. An elevated Cho/NAA or Cho/Cr ratio is therefore a hallmark of tumor recurrence. Radiation necrosis, on the other hand, is defined by widespread cell death and membrane breakdown, typically showing a pan-metabolite decrease, with particularly low Cho levels and often elevated peaks of lipids and lactate, which are byproducts of anaerobic metabolism and necrosis [40]. The meta-analysis by Chuang et al. confirmed the high diagnostic value of these ratios, reporting that the pooled difference in means for the Cho/Cr ratio was significantly higher in recurrence than necrosis (0.77, 95% CI = 0.57 to 0.98, p < 0.001) [39]. Similarly, the Cho/NAA ratio was also significantly elevated in recurrent tumors (1.02, 95% CI = 0.03 to 2.00, p = 0.044) [46]. In fact, among the various imaging modalities studied for this purpose, MRS demonstrated the highest pooled sensitivity at 90.7% [46].

5.3. Amino Acid PET: Exploiting Tumor Metabolism for Superior Diagnostic Clarity

Positron Emission Tomography (PET) provides functional information by imaging the uptake of radiolabeled tracers. While 18F-Fluorodeoxyglucose (18F-FDG) is the most common PET tracer in oncology, its utility for intracranial imaging is limited. The underlying mechanism of 18F-FDG uptake is glucose metabolism, which is physiologically high in normal brain gray matter. This high background activity can obscure the signal from metastatic lesions, resulting in a low tumor-to-background contrast and reduced diagnostic sensitivity [40].
To overcome this limitation, amino acid-based PET tracers, such as [18F]fluoroethyl-L-tyrosine ([18F]FET) and [11C]methionine ([11C]MET), have been developed and are particularly valuable in neuro-oncology. The uptake mechanism of these tracers is fundamentally different and more tumor-specific within the brain. Their transport into cells is mediated by specific amino acid transporter systems, particularly the L-type amino acid transporter 1 (LAT1), which are highly upregulated in cancer cells to meet the increased demand for protein synthesis but show very low expression in normal brain tissue. This differential expression results in high tracer accumulation in tumor tissue with minimal background uptake in the surrounding healthy brain, yielding a superior tumor-to-brain contrast and enhanced lesion detection [40]. This high contrast makes amino acid PET exceptionally effective for differentiating active tumor recurrence from treatment-related changes. Studies have consistently demonstrated a high diagnostic accuracy of 80–90% for this application (Table 5). Recognizing their value, the joint EANO-ESMO guidelines recommend considering advanced MR techniques like PWI and MRS, as well as amino acid PET, for the critical task of distinguishing tumor progression from treatment-related effects [38].

6. Guideline-Informed Therapeutic Strategies by Primary Malignancy

The management of brain metastases has evolved beyond purely local control to a personalized approach that is intimately tied to the biology of the primary tumor. In the era of precision oncology, the identification of specific molecular targets—such as driver mutations or overexpression of receptors—or the tumor’s immune status now dictates the initial systemic strategy. This section details the current, guideline-informed approaches for the three most common primary malignancies that metastasize to the brain: Non-Small Cell Lung Cancer (NSCLC), Breast Cancer, and Melanoma. For each, we outline how the availability of highly effective, CNS-penetrant systemic therapies has fundamentally shifted the treatment paradigm, often allowing for the prioritization of targeted therapy or immunotherapy upfront, with local treatments reserved for cases of primary resistance or progression.

6.1. Non-Small Cell Lung Cancer (NSCLC)

NSCLC is the most common source of brain metastases [16,19,20]. Treatment is increasingly stratified by the presence of targetable driver mutations.
EGFR-Mutant NSCLC: Patients with activating mutations in the epidermal growth factor receptor (EGFR) gene are highly sensitive to EGFR tyrosine kinase inhibitors (TKIs). While first-generation TKIs like erlotinib showed some intracranial activity, third-generation inhibitors, particularly osimertinib, have demonstrated superior CNS penetration and high intracranial response rates. For patients with asymptomatic EGFR-mutant NSCLC brain metastases, guidelines now recommend upfront treatment with osimertinib, with local therapy reserved for progression [19].
ALK-Rearranged NSCLC: Similarly to EGFR-mutant disease, NSCLC harboring an anaplastic lymphoma kinase (ALK) rearrangement responds to ALK inhibitors. Newer generation agents (e.g., alectinib, brigatinib, lorlatinib) were specifically designed for improved CNS penetration and have shown excellent intracranial efficacy, making them the preferred upfront treatment for patients with asymptomatic ALK-positive brain metastases [19].
NSCLC without Actionable Driver Mutations: For patients without targetable mutations, immunotherapy with immune checkpoint inhibitors (ICIs) targeting the PD-1/PD-L1 pathway has become a standard of care. ICIs, either as monotherapy (for PD-L1 high expressors) or in combination with chemotherapy, have demonstrated intracranial activity. There is also growing evidence for a synergistic effect when ICIs are combined with SRS, where focused radiation may enhance the systemic anti-tumor immune response [47,48].

6.2. Breast Cancer

Breast cancer is the second most common cause of brain metastases [40]. Management is dictated by the tumor’s hormone receptor (HR) and human epidermal growth factor receptor 2 (HER2) status.
HER2-Positive Breast Cancer: This subtype has a high propensity for CNS metastasis but has also seen the most significant therapeutic advances. The development of novel, CNS-penetrant anti-HER2 therapies has been transformative. The HER2CLIMB trial demonstrated that the addition of the TKI tucatinib to trastuzumab and capecitabine resulted in a significant improvement in overall survival and intracranial progression-free survival specifically for patients with active brain metastases [49,50]. Other agents like neratinib and pyrotinib have also shown substantial intracranial response rates. Consequently, ESMO guidelines support the use of these CNS-active systemic therapies to control intracranial disease and delay or avoid the need for WBRT [51].
Triple-Negative Breast Cancer (TNBC): For patients with PD-L1-positive advanced TNBC, the combination of immunotherapy (atezolizumab or pembrolizumab) with chemotherapy is an approved first-line therapy that has shown survival benefits, although specific data for its efficacy in brain metastases are still emerging [52,53].
HR-Positive/HER2-Negative Breast Cancer: This is the most common breast cancer subtype. Systemic management for metastatic disease typically involves endocrine therapy combined with targeted agents like CDK4/6 inhibitors. While effective systemically, data on the intracranial efficacy of these combinations are less mature compared to the HER2-positive setting [54].

6.3. Melanoma

Melanoma has one of the highest rates of metastasis to the brain, and these lesions are often hemorrhagic and historically associated with a very poor prognosis [55,56]. The advent of targeted therapy and immunotherapy has revolutionized outcomes.
BRAF V600-Mutant Melanoma: For the approximately 50% of melanoma patients with a BRAF V600 mutation, the combination of a BRAF inhibitor (e.g., dabrafenib) and a MEK inhibitor (e.g., trametinib) can induce rapid and high intracranial response rates [56]. This is often the preferred initial approach for symptomatic patients who require a rapid tumor response to alleviate neurological symptoms.
Immunotherapy: Immune checkpoint inhibitors have produced the most durable responses. Dual-agent immunotherapy with the combination of nivolumab (anti-PD-1) and ipilimumab (anti-CTLA-4) has achieved significantly higher and more durable intracranial response rates compared to monotherapy or historical treatments. Based on this robust evidence, combination immunotherapy is the preferred first-line treatment for patients with asymptomatic melanoma brain metastases, regardless of BRAF status, according to major clinical practice guidelines [7,55,56].
The management of melanoma brain metastases particularly highlights the sophisticated and dynamic interplay between local and systemic therapies. While upfront immunotherapy is preferred for asymptomatic patients, this is not the case for those with significant neurological symptoms requiring high-dose corticosteroids. Steroids are used to control peritumoral edema but are also immunosuppressive and can blunt the efficacy of ICIs. Therefore, for symptomatic patients, local therapy (SRS or surgery) is often employed first as a “bridging” strategy. By rapidly controlling the symptomatic lesion and allowing for the tapering of steroids, local therapy enables the safe and effective administration of subsequent immunotherapy [7]. This demonstrates that the timing and sequencing of treatments are as critical as the choice of therapy itself, underscoring the necessity for proactive, multidisciplinary tumor board discussions to choreograph these complex treatment plans for brain metastases from NSCLC, breast cancer, and melanoma, (Table 6) [7].

7. Discussion

7.1. Horizons

The management of brain metastases has undergone a paradigm shift, moving from a historically palliative approach centered on WBRT to a highly personalized, multidisciplinary strategy focused on long-term disease control and quality of life. This evolution is built upon a deeper understanding of tumor biology, technological advancements in imaging and radiation oncology, and the development of transformative systemic therapies.

7.2. Synthesis: The Four Pillars of Modern Brain Metastasis Management

The modern management of brain metastases rests on four interconnected pillars:
Molecular Stratification: Treatment decisions are no longer based solely on the number and location of lesions but are fundamentally driven by the primary tumor’s genetic and molecular profile. The identification of targetable driver mutations (e.g., EGFR, ALK, HER2, BRAF) dictates the use of specific, highly effective systemic therapies [19].
Advanced Imaging for Precision Diagnostics: The role of imaging has expanded beyond anatomical detection. Advanced functional and metabolic techniques like PWI, MRS, and amino acid PET are now essential tools for addressing the critical post-treatment dilemma of differentiating tumor recurrence from necrosis, thereby guiding subsequent therapeutic decisions with greater precision [19].
Focal Local Therapy to Preserve Cognition: There is a clear and guideline-supported shift away from WBRT toward focal therapies like SRS for patients with limited intracranial disease. This prioritization reflects a fundamental goal of modern neuro-oncology: to achieve durable local control while minimizing the neurocognitive toxicity that can severely impair a patient’s quality of life [7,57].
CNS-Active Systemic Agents: The success of targeted therapies and immunotherapies in achieving significant intracranial responses has transformed the prognosis for many patients. These agents have introduced a new strategy of using systemic treatment upfront, potentially deferring or avoiding the need for local brain-directed therapies altogether [7,19,20,57].
On the other hand, the dilemmas between radiation-induced brain necrosis and recurrent brain metastasis with associated edema remain difficult to resolve, even with conventional advanced imaging techniques. Integrated PET/MRI has emerged as a synergistic, multiparametric solution that markedly improves diagnostic accuracy by combining metabolic PET data with the structural and physiological insights of MRI. For instance, MRI-derived relative cerebral blood volume (rCBV) tends to be high in tumor recurrence due to increased neovascularization and perfusion, whereas rCBV is typically low or normal in radiation necrosis, where vascular injury and reduced blood flow predominate. Likewise, PET metabolic indices such as the standardized uptake value ratio (e.g., FET T/Wm ratio) are elevated in recurrent tumor owing to increased amino acid transport and cellular proliferation, but low in radiation necrosis, where the tissue is largely inflammatory with minimal metabolic activity [58,59]. In addition, specialized MRI techniques such as Treatment Response Assessment Maps (TRAM) further enhance differentiation accuracy. TRAM imaging is performed by acquiring early (~5 min) and delayed (60–105 min) post-contrast T1-weighted sequences, followed by subtraction to generate maps that highlight areas retaining contrast over time. Persisting enhancement on delayed images is characteristic of viable tumor, while treatment-related necrosis generally demonstrates minimal residual contrast [60].

7.3. Unresolved Questions and Clinical Controversies

Despite significant progress, several key questions and controversies remain at the forefront of clinical research.
Optimal Sequencing of SRS and Immunotherapy: The combination of SRS and ICIs is a “double-edged sword.” Preclinical and clinical data suggest a synergistic relationship, where focused radiation can induce immunogenic cell death, release tumor antigens, and potentially create a systemic “abscopal effect,” enhancing the efficacy of immunotherapy. However, this combination is also associated with a significantly increased risk of radiation necrosis, which can cause substantial neurological morbidity [61]. The optimal timing—whether SRS and ICIs should be given concurrently or sequentially—is a major unresolved question and the subject of ongoing clinical trials.
Redefining “Oligometastatic” Disease: The traditional definition of oligometastatic disease treatable with SRS has been based on a simple numerical cutoff (e.g., 1–4 lesions). However, this definition is being challenged. Emerging evidence suggests that the total intracranial tumor volume may be a more important prognostic factor than the absolute number of lesions. Recent trials have shown that SRS can be a safe and effective option for patients with a higher number of metastases (e.g., 5–15), provided the cumulative volume is manageable [62]. The field is moving toward a more nuanced, volume-based definition of which patients are suitable candidates for focal therapy.
Optimal Management of Symptomatic Radiation Necrosis: While advanced imaging has improved the diagnosis of radiation necrosis, its optimal management remains unclear. Current options range from conservative approaches like observation and corticosteroids to more active treatments like bevacizumab (which targets VEGF, a key driver of edema and vascular permeability in necrosis) or even surgical resection for large, symptomatic necrotic masses [63]. There is a lack of high-level, comparative evidence to guide the selection and sequencing of these interventions, representing a critical unmet need.
Variability in imaging acquisition protocols and reconstruction algorithms across different centers: despite the growing promise of radiomics and artificial intelligence in neuro-oncology, an important limitation lies in the variability of imaging acquisition protocols and reconstruction algorithms across different centers. Differences in scanner models, magnetic field strengths, pulse sequences, contrast administration timing, and post-processing pipelines can lead to substantial variation in the extracted radiomic features, ultimately affecting model training, performance, and reproducibility. These inconsistencies pose significant challenges for cross-institutional validation and hinder the generalizability of machine learning tools into real-world clinical practice. Continued efforts toward imaging standardization, implementation of harmonization techniques, and prospective multicenter validation are therefore essential to ensure that imaging-based biomarkers can be reliably integrated into guideline-directed management strategies for patients with brain metastases.

7.4. Concrete Research Challenges

The field continues to evolve rapidly, with several promising areas of research poised to further refine patient care.
Novel Therapeutics and Delivery Systems: The development of new systemic agents with even better CNS penetration and novel mechanisms of action remains a high priority. Additionally, innovative drug delivery strategies, such as nanoparticle-based carriers or focused ultrasound to transiently open the BBB, may enhance the efficacy of both existing and new therapies.
Liquid Biopsy for CNS Monitoring: The analysis of circulating tumor DNA (ctDNA) from cerebrospinal fluid (CSF) is emerging as a powerful, minimally invasive tool. CSF liquid biopsy has the potential to provide real-time molecular profiling of intracranial tumors, identify mechanisms of resistance, and monitor treatment response without the need for high-risk brain biopsies [64]. While not yet standard of care, it holds immense promise for future precision medicine in neuro-oncology.
Radiomics and Artificial Intelligence (AI): The application of AI and machine learning algorithms to medical imaging data is a rapidly advancing frontier. Radiomics involves the high-throughput extraction of quantitative features from standard clinical images (e.g., MRI) to create predictive models. These AI-driven tools could potentially identify subtle imaging biomarkers—imperceptible to the human eye—that predict treatment response, the risk of developing radiation necrosis, and overall prognosis, enabling an even deeper level of treatment personalization [65].

8. Conclusions

The management of brain metastases has been fundamentally transformed from a palliative-focused discipline to one of active, personalized, and often long-term disease control. This evolution is driven by the synergistic integration of advanced imaging modalities and precision-based therapies. Functional and metabolic imaging techniques such as PWI, MRS, and amino acid PET have become indispensable for accurately differentiating disease progression from treatment effects, thereby guiding more precise and timely clinical decisions. Therapeutically, the judicious use of focal local therapies like SRS, combined with a growing arsenal of molecularly targeted and immune-based systemic agents, offers improved outcomes while minimizing treatment-related toxicity. The evidence strongly supports a multidisciplinary, guideline-directed approach where treatment is tailored to the primary cancer’s molecular profile, the patient’s overall condition, and the intracranial disease burden. Continued research is essential to refine imaging biomarkers, validate novel therapeutic combinations, and develop evidence-based algorithms that further optimize care and enhance both the duration and quality of life for the growing population of patients living with brain metastases.

Author Contributions

Conceptualization, K.R., W.S. and K.R.; methodology, W.S.; software, W.S.; validation, K.R., R.N., W.N., M.A.A., A.S., E.E.D.A.Z., F.A.G., A.A.J., M.A., A.Y. and W.S.; formal analysis, K.R., R.N., W.N., M.A.A., A.S., E.E.D.A.Z., F.A.G., A.A.J., M.A., A.Y. and W.S.; investigation, K.R., R.N., W.N., M.A.A., A.S., E.E.D.A.Z., F.A.G., A.A.J., M.A., A.Y. and W.S.; resources, K.R., R.N., W.N., M.A.A., A.S., E.E.D.A.Z., F.A.G., A.A.J., M.A., A.Y. and W.S.; data curation, W.S.; writing—original draft preparation, K.R., R.N., W.N., M.A.A., A.S., E.E.D.A.Z., F.A.G., A.A.J., M.A., A.Y. and W.S.; writing—review and editing, K.R., R.N., W.N., M.A.A., A.S., E.E.D.A.Z., F.A.G., A.A.J., M.A., A.Y. and W.S.; visualization, K.R., R.N., W.N., M.A.A., A.S., E.E.D.A.Z., F.A.G., A.A.J., M.A., A.Y. and W.S.; supervision, W.S.; project ad-ministration, W.S.; funding acquisition, W.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Institutional Review Board of Soroka Medical Center. The study was approved by the Institutional Review Board of Soroka Medical Center (approval no. 0329; on 14 January 2025).

Informed Consent Statement

Patient consent was waived due to the retrospective nature of the study, which posed no risk to the rights or welfare of the participants and could not have been feasibly conducted without the waiver.

Data Availability Statement

The original contributions presented in this study are included in the article. Further inquiries can be directed to the corresponding author.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
18F-FDG18F-Fluorodeoxyglucose
ALKAnaplastic Lymphoma Kinase
ASLArterial Spin Labeling
BBBBlood–Brain Barrier
BCBMBreast Cancer Brain Metastasis
BMBrain Metastases
CBFCerebral Blood Flow
CBVCerebral Blood Volume
ChoCholine
CNSCentral Nervous System
CNS-PFSCentral Nervous System Progression-Free Survival
CrCreatine
DSCDynamic Susceptibility Contrast
EGFREpidermal Growth Factor Receptor
fMRIFunctional Magnetic Resonance Imaging
HER2Human Epidermal Growth Factor Receptor 2
ICIsImmune Checkpoint Inhibitors
iORRIntracranial Objective Response Rate
LINACLinear Accelerator
MBMMelanoma Brain Metastases
MRIMagnetic Resonance Imaging
MRSMagnetic Resonance Spectroscopy
MTTMean Transit Time
NAAN-acetyl Aspartate
NSCLCNon-Small Cell Lung Cancer
ORRObjective Response Rate
OSOverall Survival
PETPositron Emission Tomography
PFSProgression-Free Survival
PWIPerfusion-Weighted Imaging
rCBVRelative Cerebral Blood Volume
RTRadiotherapy
SRSStereotactic Radiosurgery
TKITyrosine Kinase Inhibitor
TKIsTyrosine Kinase Inhibitors
WBRTWhole-Brain Radiation Therapy

References

  1. Teixeira Loiola de Alencar, V.; Guedes Camandaroba, M.P.; Pirolli, R.; Fogassa, C.A.Z.; Cordeiro de Lima, V.C. Immunotherapy as Single Treatment for Patients with NSCLC with Brain Metastases: A Systematic Review and Meta-Analysis-the META-L-BRAIN Study. J. Thorac. Oncol. 2021, 16, 1379–1391. [Google Scholar] [CrossRef] [PubMed]
  2. Kaufmann, T.J.; Smits, M.; Boxerman, J.; Huang, R.; Barboriak, D.P.; Weller, M.; Chung, C.; Tsien, C.; Brown, P.D.; Shankar, L.; et al. Consensus recommendations for a standardized brain tumor imaging protocol for clinical trials in brain metastases. Neuro Oncol. 2020, 22, 757–772. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  3. Mehta, M.P.; Paleologos, N.A.; Mikkelsen, T.; Robinson, P.D.; Ammirati, M.; Andrews, D.W.; Asher, A.L.; Burri, S.H.; Cobbs, C.S.; Gaspar, L.E.; et al. The role of chemotherapy in the management of newly diagnosed brain metastases: A systematic review and evidence-based clinical practice guideline. J. Neurooncol. 2010, 96, 71–83. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  4. Horbinski, C.; Nabors, L.B.; Portnow, J.; Baehring, J.; Bhatia, A.; Bloch, O.; Brem, S.; Butowski, N.; Cannon, D.M.; Chao, S.; et al. NCCN Guidelines® Insights: Central Nervous System Cancers, Version 2.2022. J. Natl. Compr. Cancer Netw. 2023, 21, 12–20. [Google Scholar] [CrossRef] [PubMed]
  5. Porte, M.; Massard, C. New therapies for brain metastases: An update. Curr. Opin. Oncol. 2025, 37, 611–617. [Google Scholar] [CrossRef] [PubMed]
  6. Vogelbaum, M.A.; Brown, P.D.; Messersmith, H.; Brastianos, P.K.; Burri, S.; Cahill, D.; Dunn, I.F.; Gaspar, L.E.; Gatson, N.T.N.; Gondi, V.; et al. Treatment for Brain Metastases: ASCO-SNO-ASTRO Guideline. J. Clin. Oncol. 2022, 40, 492–516, Erratum in J. Clin. Oncol. 2022, 40, 1392. https://doi.org/10.1200/JCO.22.00593. [Google Scholar] [CrossRef] [PubMed]
  7. Aseel, A.; McCarthy, P.; Mohammed, A. Brain magnetic resonance spectroscopy to differentiate recurrent neoplasm from radiation necrosis: A systematic review and meta-analysis. J. Neuroimaging 2023, 33, 189–201. [Google Scholar] [CrossRef] [PubMed]
  8. Grenzelia, M.; Zygogianni, A.; Grapsa, D.; Maragkoudakis, E.; Fyta, E.; Charpidou, A.; Syrigos, K.; Mpakakos, P. Limited Cerebral Metastases in NSCLC: A Literature Review of SRS Versus Whole-brain Radiotherapy. Cancer Diagn. Progn. 2022, 2, 609–619. [Google Scholar] [CrossRef]
  9. Almeida, N.D.; Kuo, C.; Schrand, T.V.; Rupp, J.; Madhugiri, V.S.; Goulenko, V.; Shekher, R.; Shah, C.; Prasad, D. Stereotactic Radiosurgery for Intracranial Breast Metastases: A Systematic Review and Meta-Analysis. Cancers 2024, 16, 3551. [Google Scholar] [CrossRef]
  10. Hatiboglu, M.A.; Wildrick, D.M.; Sawaya, R. The role of surgical resection in patients with brain metastases. Ecancermedicalscience 2013, 7, 308. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  11. Altieri, R.; Corvino, S.; La Rocca, G.; Cofano, F.; Melcarne, A.; Garbossa, D.; Barbarisi, M. Clinical Implication of Brain Metastases En-Bloc Resection: Surgical Technique Description and Literature Review. J. Pers. Med. 2024, 14, 1110. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  12. Sas-Korczynska, B.; Rucinska, M. WBRT for brain metastases from non-small cell lung cancer: For whom and when?—Contemporary point of view. J. Thorac. Dis. 2021, 13, 3246–3257. [Google Scholar] [CrossRef] [PubMed]
  13. Lagerwaard, F.J.; van der Hoorn, E.A.; Verbakel, W.F.; Haasbeek, C.J.; Slotman, B.J.; Senan, S. Whole-brain radiotherapy with simultaneous integrated boost to multiple brain metastases using volumetric modulated arc therapy. Int. J. Radiat. Oncol. Biol. Phys. 2009, 75, 253–259. [Google Scholar] [CrossRef] [PubMed]
  14. Latorzeff, I.; Antoni, D.; Josset, S.; Noël, G.; Tallet-Richard, A. Radiation therapy for brain metastases. Cancer Radiother. 2022, 26, 129–136. [Google Scholar] [CrossRef] [PubMed]
  15. Garsa, A.; Jang, J.K.; Baxi, S.; Chen, C.; Akinniranye, O.; Hall, O.; Larkin, J.; Motala, A.; Hempel, S. Radiation Therapy for Brain Metastases: A Systematic Review. Pract. Radiat. Oncol. 2021, 11, 354–365. [Google Scholar] [CrossRef] [PubMed]
  16. Mathis, N.J.; Wijetunga, N.A.; Imber, B.S.; Pike, L.R.G.; Yang, J.T. Recent Advances and Applications of Radiation Therapy for Brain Metastases. Curr. Oncol. Rep. 2022, 24, 335–342. [Google Scholar] [CrossRef] [PubMed]
  17. Liu, L.; Chen, W.; Zhang, R.; Wang, Y.; Liu, P.; Lian, X.; Zhang, F.; Wang, Y.; Ma, W. Radiotherapy in combination with systemic therapies for brain metastases: Current status and progress. Cancer Biol. Med. 2020, 17, 910–922. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  18. Yang, S.; Xiao, J.; Liu, Q.; Zhang, Y.; Bi, N.; Huang, X.; Chen, X.; Wang, K.; Ma, Y.; Deng, L.; et al. The Sequence of Intracranial Radiotherapy and Systemic Treatment with Tyrosine Kinase Inhibitors for Gene-Driven Non-Small Cell Lung Cancer Brain Metastases in the Targeted Treatment Era: A 10-Year Single-Center Experience. Front. Oncol. 2021, 11, 732883. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  19. Shalata, W.; Naamneh, R.; Najjar, W.; Abu Amna, M.; Asla, M.; Agbarya, A.; Brenner, R.; Abu Jama, A.; Abu Yasin, N.; Abu Juda, M.; et al. Efficacy of Tyrosine Kinase Inhibitors in ALK and EGFR-Mutated Non-Small Cell Lung Cancer with Brain Metastases. Med. Sci. 2025, 13, 200. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  20. Shalata, W.; Naamneh, R.; Najjar, W.; Asla, M.; Abu Gameh, A.; Abu Amna, M.; Saiegh, L.; Agbarya, A. Current and Emerging Therapeutic Strategies for Limited- and Extensive-Stage Small-Cell Lung Cancer. Med. Sci. 2025, 13, 142. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  21. Verduin, M.; Zindler, J.D.; Martinussen, H.M.; Jansen, R.L.; Croes, S.; Hendriks, L.E.; Eekers, D.B.; Hoeben, A. Use of Systemic Therapy Concurrent with Cranial Radiotherapy for Cerebral Metastases of Solid Tumors. Oncologist 2017, 22, 222–235. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  22. Thompson, J.F.; Williams, G.J.; Hong, A.M. Radiation therapy for melanoma brain metastases: A systematic review. Radiol. Oncol. 2022, 56, 267–284. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  23. Tonse, R.; Tom, M.C.; Mehta, M.P.; Ahluwalia, M.S.; Kotecha, R. Integration of Systemic Therapy and Stereotactic Radiosurgery for Brain Metastases. Cancers 2021, 13, 3682. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  24. Pomeranz Krummel, D.A.; Nasti, T.H.; Izar, B.; Press, R.H.; Xu, M.; Lowder, L.; Kallay, L.; Rupji, M.; Rosen, H.; Su, J.; et al. Impact of Sequencing Radiation Therapy and Immune Checkpoint Inhibitors in the Treatment of Melanoma Brain Metastases. Int. J. Radiat. Oncol. Biol. Phys. 2020, 108, 157–163. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  25. Cleveland Clinic. MRI (Magnetic Resonance Imaging): What it is & results [Internet]. Cleveland Clinic. Available online: https://my.clevelandclinic.org/health/diagnostics/4876-magnetic-resonance-imaging-mri (accessed on 17 October 2025).
  26. Moffitt Cancer Center. Brain Tumor Diagnosis—MRI. Moffitt Cancer Center. Available online: https://www.moffitt.org/cancers/brain-tumor/diagnosis/mri/ (accessed on 17 October 2025).
  27. Mayfield Clinic. Magnetic Resonance Spectroscopy (MR Spectroscopy). Mayfield Clinic. Available online: https://mayfieldclinic.com/pe-mrspectroscopy.htm (accessed on 17 October 2025).
  28. International Brain Tumor Center and Brain Network Institute. Neuroimaging for Brain Tumors. Available online: https://www.ivybraintumorcenter.org/brain-tumor-care/brain-tumor-treatments/neuroimaging-for-brain-tumors (accessed on 17 October 2025).
  29. Sutter Health Care. How PET Scans Work—What to Expect During a PET Scan. Available online: https://stanfordhealthcare.org/medical-tests/p/pet-scan/what-to-expect.html (accessed on 17 October 2025).
  30. The Preston Robert Tisch Brain Tumor Center. What Is a PET Scan? [Internet]. Duke University. Available online: https://tischbraintumorcenter.duke.edu/blog/what-pet-scan (accessed on 17 October 2025).
  31. Kapoor, M.; Heston, T.F.; Kasi, A. PET Scanning; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar]
  32. Werner, J.M.; Lohmann, P.; Fink, G.R.; Langen, K.J.; Galldiks, N. Current landscape and emerging fields of PET imaging in patients with brain tumors. Molecules 2020, 25, 1471. [Google Scholar] [CrossRef]
  33. Schlürmann, T.; Waschulzik, B.; Combs, S.; Gempt, J.; Wiestler, B.; Weber, W.; Yakushev, I. Utility of amino acid PET in the differential diagnosis of recurrent brain metastases and treatment-related changes: A meta-analysis. J. Nucl. Med. 2023, 64, 816–821. [Google Scholar] [CrossRef]
  34. Lee, S. Perfusion-Weighted Imaging in Brain Tumor Surgery [Internet]. Number Analytics. Available online: https://www.numberanalytics.com/blog/perfusion-weighted-imaging-guide (accessed on 17 October 2025).
  35. Svolos, P.; Kousi, E.; Kapsalaki, E.; Theodorou, K.; Fezoulidis, I.; Kappas, C.; Tsougos, I. The role of diffusion and perfusion weighted imaging in the differential diagnosis of cerebral tumors: A review and future perspectives. Cancer Imaging 2014, 14, 20. [Google Scholar] [CrossRef] [PubMed]
  36. Petrella, J.R.; Provenzale, J.M. MR perfusion imaging of the brain. Am. J. Roentgenol. 2000, 175, 207–219. [Google Scholar] [CrossRef] [PubMed]
  37. Schack, A.; Aunan-Diop, J.S.; Gerhardt, F.A.; Pedersen, C.B.; Halle, B.; Kofoed, M.S.; Markovic, L.; Wirenfeldt, M.; Poulsen, F.R. Evaluating the efficacy of perfusion MRI and conventional MRI in distinguishing recurrent cerebral metastasis from brain radiation necrosis. Brain Sci. 2024, 14, 321. [Google Scholar] [CrossRef]
  38. Cho, S.K.; Na, D.G.; Ryoo, J.W.; Roh, H.G.; Moon, C.H.; Byun, H.S.; Kim, J.H. Perfusion MR imaging: Clinical utility for the differential diagnosis of various brain tumors. Korean J. Radiol. 2002, 3, 171–179. [Google Scholar] [CrossRef]
  39. Chuang, M.T.; Liu, Y.S.; Tsai, Y.S.; Chen, Y.C.; Wang, C.K. Differentiating radiation-induced necrosis from recurrent brain tumor using MR perfusion and spectroscopy: A meta-analysis. PLoS ONE 2016, 11, e0141438. [Google Scholar] [CrossRef] [PubMed]
  40. Boujraf, A.H.S. Magnetic resonance spectroscopy in the diagnosis and follow-up of brain tumors. J. Biomed. Sci. Eng. 2012, 5, 853–861. [Google Scholar] [CrossRef]
  41. Verma, N.; Cowperthwaite, M.C.; Burnett, M.G.; Markey, M.K. Differentiating tumor recurrence from treatment necrosis: A review of neuro-oncologic imaging strategies. Neuro Oncol. 2013, 15, 515–534. [Google Scholar] [CrossRef]
  42. Jajodia, A.; Goel, V.; Goyal, J.; Patnaik, N.; Khoda, J.; Pasricha, S.; Gairola, M. Combined Diagnostic Accuracy of Diffusion and Perfusion MR Imaging to Differentiate Radiation-Induced Necrosis from Recurrence in Glioblastoma. Diagnostics 2022, 12, 718. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  43. Li, Y.; Jin, G.; Su, D. Comparison of Gadolinium-enhanced MRI and 18FDG PET/PET-CT for the diagnosis of brain metastases in lung cancer patients: A meta-analysis of 5 prospective studies. Oncotarget 2017, 8, 35743–35749. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  44. Henssen, D.; Leijten, L.; Meijer, F.J.A.; van der Kolk, A.; Arens, A.I.J.; Ter Laan, M.; Smeenk, R.J.; Gijtenbeek, A.; van de Giessen, E.M.; Tolboom, N.; et al. Head-To-Head Comparison of PET and Perfusion Weighted MRI Techniques to Distinguish Treatment Related Abnormalities from Tumor Progression in Glioma. Cancers 2023, 15, 2631. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  45. Kim, D.H. Longitudinal Analysis of Amyloid PET and Brain MRI for Predicting Conversion from Mild Cognitive Impairment to Alzheimer’s Disease: Findings from the ADNI Cohort. Tomography 2025, 11, 37. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  46. Ghia, A.J.; Tward, J.D.; Anker, C.J.; Boucher, K.M.; Jensen, R.L.; Shrieve, D.C. Radiosurgery for melanoma brain metastases: The impact of hemorrhage on local control. J. Radiosurg. SBRT 2014, 3, 43–50. [Google Scholar] [PubMed]
  47. Lazaro, T.; Brastianos, P.K. Immunotherapy and targeted therapy in brain metastases: Emerging options in precision medicine. CNS Oncol. 2017, 6, 139–151. [Google Scholar] [CrossRef] [PubMed]
  48. Ranjan, T.; Podder, V.; Margolin, K.; Velcheti, V.; Maharaj, A.; Ahluwalia, M.S. Immune Checkpoint Inhibitors in the Management of Brain Metastases from Non-Small Cell Lung Cancer: A Comprehensive Review of Current Trials, Guidelines and Future Directions. Cancers 2024, 16, 3388. [Google Scholar] [CrossRef]
  49. Luo, J.; Ren, A.; Si, D.; Yang, J.; Xu, D.; Li, N. Drug treatment of breast cancer brain metastases: Progress and challenges. Discov. Oncol. 2025, 16, 1025. [Google Scholar] [CrossRef]
  50. Curigliano, G.; Mueller, V.; Borges, V.; Hamilton, E.; Hurvitz, S.; Loi, S.; Murthy, R.; Okines, A.; Paplomata, E.; Cameron, D.; et al. Tucatinib versus placebo added to trastuzumab and capecitabine for patients with pretreated HER2+ metastatic breast cancer with and without brain metastases (HER2CLIMB): Final overall survival analysis. Annu. Oncol. 2022, 33, 321–329, Erratum in Annu. Oncol. 2023, 34, 630. https://doi.org/10.1016/j.annonc.2022.12.005. [Google Scholar] [CrossRef] [PubMed]
  51. Ferreri, A.J.M.; Illerhaus, G.; Doorduijn, J.K.; Auer, D.P.; Bromberg, J.E.C.; Calimeri, T.; Cwynarski, K.; Fox, C.P.; Hoang-Xuan, K.; Malaise, D.; et al. Primary central nervous system lymphomas: EHA-ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Annu. Oncol. 2024, 35, 491–507. [Google Scholar] [CrossRef] [PubMed]
  52. Im, S.A.; Cortes, J.; Cescon, D.W.; Yusof, M.M.; Iwata, H.; Masuda, N.; Takano, T.; Huang, C.S.; Chung, C.F.; Tsugawa, K.; et al. Results from the randomized KEYNOTE-355 study of pembrolizumab plus chemotherapy for Asian patients with advanced TNBC. NPJ Breast Cancer 2024, 10, 79. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  53. Gion, M.; Blancas, I.; Cortez-Castedo, P.; Cortés-Salgado, A.; Marmé, F.; Blanch, S.; Morales, S.; Díaz, N.; Calvo-Plaza, I.; Recalde, S.; et al. Atezolizumab plus paclitaxel and bevacizumab as first-line treatment of advanced triple-negative breast cancer: The ATRACTIB phase 2 trial. Nat. Med. 2025, 31, 2746–2754. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  54. Walsh, E.M.; Smith, K.L.; Stearns, V. Management of hormone receptor-positive, HER2-negative early breast cancer. Semin. Oncol. 2020, 47, 187–200. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  55. Zheng, S.; Lin, Z.; Zhang, R.; Cheng, Z.; Li, K.; Gu, C.; Chen, Y.; Lin, J. Progress in immunotherapy for brain metastatic melanoma. Front. Oncol. 2024, 14, 1485532. [Google Scholar] [CrossRef] [PubMed]
  56. Tan, X.L.; Le, A.; Scherrer, E.; Tang, H.; Kiehl, N.; Han, J.; Jiang, R.; Diede, S.J.; Shui, I.M. Systematic literature review and meta-analysis of clinical outcomes and prognostic factors for melanoma brain metastases. Front. Oncol. 2022, 12, 1025664. [Google Scholar] [CrossRef]
  57. Smith, E.J.; Naik, A.; Shaffer, A.; Goel, M.; Krist, D.T.; Liang, E.; Furey, C.G.; Miller, W.K.; Lawton, M.T.; Barnett, D.H.; et al. Differentiating radiation necrosis from tumor recurrence: A systematic review and diagnostic meta-analysis comparing imaging modalities. J. Neurooncol. 2023, 162, 15–23. [Google Scholar] [CrossRef]
  58. Hojjati, M.; Badve, C.; Garg, V.; Tatsuoka, C.; Rogers, L.; Sloan, A.; Faulhaber, P.; Ros, P.R.; Wolansky, L.J. Role of FDG-PET/MRI, FDG-PET/CT, and Dynamic Susceptibility Contrast Perfusion MRI in Differentiating Radiation Necrosis from Tumor Recurrence in Glioblastomas. J. Neuroimaging 2018, 28, 118–125. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  59. Sahu, A.; Mathew, R.; Ashtekar, R.; Dasgupta, A.; Puranik, A.; Mahajan, A.; Janu, A.; Choudhari, A.; Desai, S.; Patnam, N.G.; et al. The complementary role of MRI and FET PET in high-grade gliomas to differentiate recurrence from radionecrosis. Front. Nucl. Med. 2023, 3, 1040998. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  60. Zach, L.; Guez, D.; Last, D.; Daniels, D.; Grober, Y.; Nissim, O.; Hoffmann, C.; Nass, D.; Talianski, A.; Spiegelmann, R.; et al. Delayed contrast extravasation MRI: A new paradigm in neuro-oncology. Neuro Oncol. 2015, 17, 457–465. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  61. Janopaul-Naylor, J.R.; Shen, Y.; Qian, D.C.; Buchwald, Z.S. The Abscopal Effect: A Review of Pre-Clinical and Clinical Advances. Int. J. Mol. Sci. 2021, 22, 11061. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  62. Brown, D.R.; Lanciano, R.; Heal, C.; Hanlon, A.; Yang, J.; Feng, J.; Stanley, M.; Buonocore, R.; Okpaku, A.; Ding, W.; et al. The effect of whole-brain radiation (WBI) and Karnofsky performance status (KPS) on survival of patients receiving stereotactic radiosurgery (SRS) for second brain metastatic event. J. Radiat. Oncol. 2017, 6, 31–37. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  63. Chitoran, E.; Rotaru, V.; Stefan, D.C.; Gullo, G.; Simion, L. Blocking Tumoral Angiogenesis VEGF/VEGFR Pathway: Bevacizumab-20 Years of Therapeutic Success and Controversy. Cancers 2025, 17, 1126. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  64. Zheng, M.M.; Zhou, Q.; Chen, H.J.; Jiang, B.Y.; Tang, L.B.; Jie, G.L.; Tu, H.Y.; Yin, K.; Sun, H.; Liu, S.Y.; et al. Cerebrospinal fluid circulating tumor DNA profiling for risk stratification and matched treatment of central nervous system metastases. Nat. Med. 2025, 31, 1547–1556. [Google Scholar] [CrossRef] [PubMed]
  65. Pinto-Coelho, L. How Artificial Intelligence Is Shaping Medical Imaging Technology: A Survey of Innovations and Applications. Bioengineering 2023, 10, 1435. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
Table 1. Summary of Pivotal Trials Comparing Stereotactic Radiosurgery (SRS) and Whole-Brain Radiation Therapy (WBRT).
Table 1. Summary of Pivotal Trials Comparing Stereotactic Radiosurgery (SRS) and Whole-Brain Radiation Therapy (WBRT).
Comparison GroupLocal Control OutcomeOverall Survival OutcomeOverall Survival OutcomeKey Findings/NuancesReference
WBRT vs. WBRT + SRS BoostImproved local control with SRS boost (e.g., 1-year local failure: 100% vs. 8%; RTOG 9508: 82% vs. 71%)Inconsistent OS benefit; OS benefit for single brain metastasis (BM), RPA class I, GPA 3.5–4OS benefit only for single BM (6.5 vs. 4.9 months) and good prognosis patientsSRS boost significantly improves local control. OS benefit is subgroup-dependent, favoring patients with better prognostic factors.[8]
SRS Alone vs. WBRT + SRSWBRT + SRS significantly improves brain tumor recurrence (BTR) control (e.g., 1-year CNS recurrence: 27% vs. 73%; BTR-free survival: 7.4 vs. 21.6 months)Inconsistent OS benefit; some studies show better OS with SRS alone (e.g., 15.2 vs. 7 months); OS benefit for younger patients with SRS alone; WBRT + SRS shows OS benefit in favorable prognosis (DS-GPA 2.5–4)Inconsistent OS benefit; some studies show no difference or favor SRS alone.WBRT improves local and distant control. OS outcomes vary—SRS alone may benefit younger or lower-volume patients; combined therapy better for high-risk/favorable prognosis groups.[8]
Table 2. Overview of Key Perfusion Parameters and Their Clinical Relevance.
Table 2. Overview of Key Perfusion Parameters and Their Clinical Relevance.
ParameterPhysiological MeaningTypical Pattern in High-Grade GliomaTypical Pattern in Low-Grade GliomaTypical Pattern in LymphomaTypical Pattern in MetastasisTypical Pattern in Radiation Necrosis
Relative Cerebral Blood Volume (rCBV)Amount of blood in a given tissue volumeMarkedly IncreasedLowLowHighLow
Relative Cerebral Blood Flow (rCBF)Rate of blood flow through a tissueMarkedly IncreasedLowLowHighLow
Mean Transit Time (MTT)Average time for blood to pass through a tissueDecreasedNormal/IncreasedNormal/IncreasedDecreasedIncreased
Table 3. Characteristic Metabolite Ratios in Brain Tumors and Treatment-Related Changes.
Table 3. Characteristic Metabolite Ratios in Brain Tumors and Treatment-Related Changes.
Metabolite Ratio/PresenceNormal Brain TissueHigh-Grade GliomaLow-Grade GliomaRadiation NecrosisMeningioma
Cho/NAA RatioBaseline/LowElevatedSlightly ElevatedLow/NormalVariable
Cho/Cr RatioBaseline/NormalElevatedSlightly ElevatedLow/NormalVariable
NAA LevelsHighDecreasedModerately DecreasedVariableNormal
Abbreviations: Choline, Cho; N-acetyl aspartate, NAA; Creatine, Cr.
Table 4. Quantitative comparisons data of sensitivity, specificity, and AUC values derived from various meta-analyses and studies.
Table 4. Quantitative comparisons data of sensitivity, specificity, and AUC values derived from various meta-analyses and studies.
ModalitySensitivitySpecificityAUCNotes
Conventional MRI~77% (0.60–0.89)~99% (0.97–1.00)~0.97Data specifically for lung cancer brain metastases.
Perfusion-Weighted Imaging (PWI)~80%~86%~0.93Data for distinguishing tumor progression from radiation necrosis using relative cerebral blood volume (rCBV) threshold.
Magnetic Resonance Spectroscopy (MRS)Not widely pooledNot widely pooledNot widely pooledDescribed as having high diagnostic accuracy, especially when combined with other methods.
FDG-PET~21% (0.13–0.32)~100% (0.99–1.00)~0.98Low sensitivity for brain metastases in lung cancer, but high specificity.
[11C] MET PET89% (78–95%)72% (25–95%)Not widely pooledData for differentiating tumor progression from treatment related abnormalities.
[18F]-FET PET82% (72–90%)85% (68–94%)Not widely pooledData for differentiating tumor progression from treatment related abnormalities.
Table 5. Advanced Imaging Characteristics of Tumor Recurrence vs. Radiation Necrosis.
Table 5. Advanced Imaging Characteristics of Tumor Recurrence vs. Radiation Necrosis.
ModalityKey ParameterFinding in Tumor RecurrenceFinding in Radiation Necrosis
PWIRelative Cerebral Blood Volume (rCBV)IncreasedDecreased/Low
MRSCholine/Creatine (Cho/Cr) RatioElevatedLow/Normal
MRSCholine/NAA (Cho/NAA) RatioElevatedLow/Normal
Amino Acid PETTumor-to-Brain Uptake RatioHighLow
Table 6. Guideline-Based Systemic Therapies with Established CNS Activity.
Table 6. Guideline-Based Systemic Therapies with Established CNS Activity.
Primary CancerSubtype/BiomarkerDrug ClassRecommended Agents (Examples)Key Efficacy Data
NSCLCEGFR Mutation3rd-Gen TKIOsimertinibHigh intracranial response rate; recommended as upfront therapy for asymptomatic BM.
NSCLCALK Rearrangement2nd/3rd-Gen TKIAlectinib, Brigatinib, LorlatinibExcellent CNS penetration and intracranial efficacy; preferred upfront therapy.
Breast CancerHER2-PositiveTKITucatinib (+Trastuzumab/Capecitabine)HER2CLIMB: Significant OS and CNS-PFS benefit in patients with active BM.
Breast CancerHER2-PositiveTKINeratinib, PyrotinibPERMEATE (Pyrotinib): High CNS-ORR (74.6% in RT-naive patients).
MelanomaBRAF V600 MutationBRAF/MEK InhibitorsDabrafenib + TrametinibHigh intracranial ORR (up to 58%); used for rapid response in symptomatic patients.
MelanomaAnyDual ImmunotherapyNivolumab + IpilimumabHigh and durable intracranial response rates (~54%); preferred first-line for asymptomatic BM.
Abbreviation: NSCLC, Non-Small Cell Lung Cancer; EGFR, Epidermal Growth Factor Receptor; TKI, Tyrosine Kinase Inhibitor; ALK, Anaplastic Lymphoma Kinase; HER2, Human Epidermal Growth Factor Receptor 2; BM, Brain Metastases; CNS, Central Nervous System; OS, Overall Survival; PFS, Progression-Free Survival; CNS-PFS, Central Nervous System Progression-Free Survival; ORR, Objective Response Rate; RT, Radiotherapy.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Rouvinov, K.; Naamneh, R.; Najjar, W.; Abu Amna, M.; Soklakova, A.; Abu Zeid, E.E.D.; Abu Ghalion, F.; Abu Juma’a, A.; Asla, M.; Yakobson, A.; et al. Integrating Advanced Neuro-Oncology Imaging into Guideline-Directed Multimodal Therapy for Brain Metastases: Evaluating Comparative Treatment Effectiveness. Technologies 2025, 13, 532. https://doi.org/10.3390/technologies13110532

AMA Style

Rouvinov K, Naamneh R, Najjar W, Abu Amna M, Soklakova A, Abu Zeid EED, Abu Ghalion F, Abu Juma’a A, Asla M, Yakobson A, et al. Integrating Advanced Neuro-Oncology Imaging into Guideline-Directed Multimodal Therapy for Brain Metastases: Evaluating Comparative Treatment Effectiveness. Technologies. 2025; 13(11):532. https://doi.org/10.3390/technologies13110532

Chicago/Turabian Style

Rouvinov, Keren, Rashad Naamneh, Wenad Najjar, Mahmoud Abu Amna, Arina Soklakova, Ez El Din Abu Zeid, Fahmi Abu Ghalion, Ali Abu Juma’a, Mohnnad Asla, Alexander Yakobson, and et al. 2025. "Integrating Advanced Neuro-Oncology Imaging into Guideline-Directed Multimodal Therapy for Brain Metastases: Evaluating Comparative Treatment Effectiveness" Technologies 13, no. 11: 532. https://doi.org/10.3390/technologies13110532

APA Style

Rouvinov, K., Naamneh, R., Najjar, W., Abu Amna, M., Soklakova, A., Abu Zeid, E. E. D., Abu Ghalion, F., Abu Juma’a, A., Asla, M., Yakobson, A., & Shalata, W. (2025). Integrating Advanced Neuro-Oncology Imaging into Guideline-Directed Multimodal Therapy for Brain Metastases: Evaluating Comparative Treatment Effectiveness. Technologies, 13(11), 532. https://doi.org/10.3390/technologies13110532

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop