Next Article in Journal
Association of Systemic Pathologies on Dental, Periodontal and Orthodontic Status in Children
Previous Article in Journal
Exploring the Cellular and Molecular Landscape of Idiopathic Pulmonary Fibrosis: Integrative Multi-Omics and Single-Cell Analysis
Previous Article in Special Issue
Glioblastoma: From Pathophysiology to Novel Therapeutic Approaches
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Radiotherapy in Glioblastoma Multiforme: Evolution, Limitations, and Molecularly Guided Future

1
Department of Radiation Oncology, Hospital Universitario San Francisco de Asís, GenesisCare, 28002 Madrid, Spain
2
Department of Radiation Oncology, Hospital Universitario Vithas La Milagrosa, GenesisCare, 28010 Madrid, Spain
3
Department of Radiation Oncology, Hospital Universitario HM Sanchinarro, HM Hospitales, 28050 Madrid, Spain
4
Radiation Therapy Department, Quironsalud Madrid University Hospital, Diego de Velázquez 1, 28223 Pozuelo de Alarcón, Madrid, Spain
5
Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28670 Villaviciosa de Odón, Madrid, Spain
*
Author to whom correspondence should be addressed.
Biomedicines 2025, 13(9), 2136; https://doi.org/10.3390/biomedicines13092136
Submission received: 7 July 2025 / Revised: 24 August 2025 / Accepted: 29 August 2025 / Published: 1 September 2025
(This article belongs to the Special Issue Glioblastoma: From Pathophysiology to Novel Therapeutic Approaches)

Abstract

Glioblastoma multiforme (GBM), the most aggressive primary brain tumor in adults, has a poor prognosis due to rapid recurrence and treatment resistance. This review examines the evolution of radiotherapy (RT) for GBM management, from whole-brain RT to modern techniques like intensity-modulated RT (IMRT) and volumetric modulated arc therapy (VMAT), guided by 2023 European Society for Radiotherapy and Oncology (ESTRO)-European Association of Neuro-Oncology (EANO) and 2025 American Society for Radiation Oncology (ASTRO) recommendations. The standard Stupp protocol (60 Gy/30 fractions with temozolomide [TMZ]) improves overall survival (OS) to 14.6 months, with greater benefits in O6-methylguanine-DNA methyltransferase (MGMT)-methylated tumors (21.7 months). Tumor Treating Fields (TTFields) extend median overall survival (mOS) to 31.6 months in MGMT-methylated patients and 20.9 months overall in supratentorial GBM (EF-14 trial). However, 80–90% of recurrences occur within 2 cm of the irradiated field due to tumor infiltration and radioresistance driven by epidermal growth factor receptor (EGFR) amplification, phosphatase and tensin homolog (PTEN) mutations, cyclin-dependent kinase inhibitor 2A/B (CDKN2A/B) deletions, tumor hypoxia, and tumor stem cells. Pseudoprogression, distinguished using Response Assessment in Neuro-Oncology (RANO) criteria and positron emission tomography (PET), complicates response evaluation. Targeted therapies (e.g., bevacizumab; PARP inhibitors) and immunotherapies (e.g., pembrolizumab; oncolytic viruses), alongside advanced imaging (multiparametric magnetic resonance imaging [MRI], amino acid PET), support personalized RT. Ongoing trials evaluating reirradiation, hypofractionation, stereotactic radiosurgery, neoadjuvant therapies, proton therapy (PT), boron neutron capture therapy (BNCT), and AI-driven planning aim to enhance efficacy for GBM IDH-wildtype, but phase III trials are needed to improve survival and quality of life.

1. Introduction

Glioblastoma multiforme (GBM), the most common malignant primary brain tumor in adults, has an incidence of 3–5 cases per 100,000 people annually, with a male predominance (1.6:1) and peak incidence at 50–60 years [1,2,3,4]. According to the 2021 World Health Organization (WHO) classification, GBM is defined as an isocitrate dehydrogenase (IDH)-wild-type (IDH-wt), grade 4 astrocytoma, diagnosed by histological features (necrosis; microvascular proliferation) or molecular markers, including telomerase reverse transcriptase (TERT) promoter mutation, epidermal growth factor receptor (EGFR) amplification, or +7/−10 chromosomal alterations [5]. Most GBMs arise de novo, though a small percentage develop from lower-grade gliomas; risk factors include prior cranial radiotherapy (RT) or hereditary syndromes like Cowden, Turcot, Lynch, Li-Fraumeni, or neurofibromatosis type I [2]. Multidisciplinary treatment combines maximal safe resection followed by RT and concurrent chemotherapy (CTx), with no defined second-line therapy, necessitating personalized approaches [2,6]. Standard RT techniques, such as intensity-modulated RT (IMRT) and volumetric modulated arc therapy (VMAT) for precise dose delivery, are widely used, while experimental proton therapy (PT) and boron neutron capture therapy (BNCT) are under investigation, as detailed in Section 5.1. Despite advances, limited progress in outcomes underscores the need for innovative strategies to improve oncologic results and quality of life, as explored in Section 2, Section 3, Section 4 and Section 5. This review examines the evolution, limitations, and molecularly guided advances in RT for GBM management.

Search Strategy

To inform this review, we consulted PubMed, Scopus, and Google Scholar, focusing on clinical trials and guidelines (2023–2025) for GBM radiotherapy, using terms like ‘glioblastoma radiotherapy’, ‘GBM molecular biomarkers’, and ‘proton therapy GBM’. We prioritized English articles from 2000 to 2025, emphasizing clinical trials and guidelines, and excluded non-human studies.

2. Evolution of Radiotherapy in GBM: Foundations, Standards, and International Guidelines

GBM is characterized by rapid progression and treatment resistance, with a median overall survival (mOS) of 12–15 months and a 5-year survival rate below 10%, varying by prognostic factors such as age, functional status (Karnofsky Performance Status [KPS]), O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation, tumor location, and involvement of deep structures or functional areas [6,7,8,9]. The first-line treatment for GBM involves maximal safe resection, followed by adjuvant RT and CTx to target residual microscopic disease due to the tumor’s infiltrative nature [10,11]. Preoperative neuronavigation magnetic resonance imaging (MRI) and early postoperative contrast-enhanced MRI (within 48–72 h) are recommended, with intraoperative biopsy for diagnosis confirmation if resection is not feasible [2,10]. RT should begin 3–6 weeks post-surgery to minimize recurrence risk [10,11]. The standard protocol, established by the European Organisation for Research and Treatment of Cancer (EORTC) 26981/22981 trial, combines normofractionated RT (60 Gy in 30 fractions) with concomitant temozolomide (TMZ, 75 mg/m2/day), followed by 6 cycles of adjuvant TMZ (150–200 mg/m2/day, 5/28 days), improving overall survival (OS) from 12.1 to 14.6 months, particularly in patients with MGMT promoter methylation (Table 1) [12]. This regimen is recommended for patients ≤70 years with good functional status (Karnofsky Performance Status [KPS] ≥60) [13,14]. Extending TMZ beyond 6 cycles lacks evidence [15]. Tumor Treating Fields (TTFields), which disrupt cell division with alternating electric fields, are initiated 4–7 weeks post-radiotherapy during maintenance TMZ in selected patients (supratentorial GBM; KPS ≥60), improving mOS to 31.6 months in MGMT-methylated patients and 20.9 months overall (EF-14 trial), with minimal toxicity (mainly skin irritation), and are endorsed by National Comprehensive Cancer Network (NCCN) and American Society for Radiation Oncology (ASTRO) 2025 guidelines (Table 1) [12,16,17]. Other therapeutic innovations and systemic treatment options are discussed in the therapeutic innovations section.
Hypofractionated RT (40.05 Gy/15 fractions with TMZ, 34 Gy/10 fractions, or 25 Gy/5 fractions without TMZ) is the preferred standard of care for elderly (≥70 years) or frail patients (KPS 50–70), offering comparable overall survival (OS), significantly reduced toxicity, and enhanced quality of life, as supported by clinical trials and meta-analyses (Table 2) [18,19,20,21,22,23,24]. ASTRO 2025 and European Society for Radiotherapy and Oncology (ESTRO)-European Association of Neuro-Oncology (EANO) 2023 guidelines conditionally recommend these regimens for elderly or frail patients, while those with poor KPS (≤40) or extreme frailty may receive hypofractionated RT alone, TMZ alone (if MGMT-methylated), or palliative care to prioritize quality of life [10,11,13,14,25].
RT techniques have evolved from whole-brain RT (WBRT) in the 1970s to three-dimensional conformal RT (3D-CRT) in the 1990s, and now to IMRT and VMAT, which optimize dose delivery and spare healthy tissues. Treatment planning integrates thin-slice CT and MRI (T1 with gadolinium, T2/FLAIR) performed ≤14 days before RT for precise tumor and organ-at-risk delineation. ASTRO 2025 and ESTRO-EANO 2023 [11,25] guidelines prioritize IMRT/VMAT over 3D-CRT to reduce neurological toxicity. ESTRO-EANO 2023 recommends a single-phase approach (gross tumor volume [GTV]: surgical cavity + T1 enhancement; clinical target volume [CTV]: GTV + 15 mm; edema optional only for non-hyperintense T1 tumors), while ASTRO 2025 allows a single phase (edema optional) or two phases (initial phase with T2/FLAIR; boost without edema), with CTV of 10–20 mm and planning target volume (PTV) of 2–5 mm with image-guided RT (IGRT) (Table 3) [11,12,14,25,26,27,28,29]. A randomized trial with 245 patients with grade 3–4 gliomas compared the RTOG/NRG approach (including edema with a boost) and the EORTC approach (including edema in the initial phase), finding no significant differences in grade 3–4 toxicity (36.1% vs. 32.3%) or recurrence patterns (predominantly central), though neurocognitive outcomes were not assessed [30,31]. These findings, consistent with prior studies, support the flexibility of ASTRO 2025 and ESTRO-EANO 2023 guidelines, which allow either single-phase (edema optional) or two-phase (edema in initial phase) approaches (Table 3) [11,25].
The MD Anderson Cancer Center (MDACC) protocol, excluding edema, improved OS (17 vs. 12 months) and quality of life without increasing toxicity, suggesting smaller volumes may be beneficial (Table 3) [27].
Post-treatment follow-up includes MRI at 4 weeks, then every 2–4 months, per Response Assessment in Neuro-Oncology (RANO) criteria. Pseudoprogression, challenging to distinguish from true progression, may require advanced imaging (diffusion MRI, spectroscopy MRI, or amino acid positron emission tomography (PET)/CT), correlating findings with conventional MRI. In case of progression, a personalized approach is recommended, considering tumor characteristics (size, location, and molecular profile), initial treatment response, age, KPS, symptoms, needs, and patient preferences, with multidisciplinary evaluation of options (reoperation, reirradiation, or systemic therapy) [11,25]. These advances underscore the need for tailored RT strategies to optimize GBM treatment outcomes.

3. Limitations of Radiotherapy in GBM

RT combined with TMZ is a cornerstone of GBM treatment but faces technical, biological, and clinical limitations that reduce efficacy and lead to high recurrence and resistance rates. These barriers, summarized in Table 4, underscore the need for innovative strategies discussed later.
Technically, 90% of GBMs recur within 2 cm of the irradiated field due to diffuse infiltration [32], despite advanced techniques like IMRT, VMAT, or proton therapy (PT). High doses cause fatigue, radiation necrosis, and cognitive deficits, linked to irradiated brain volumes receiving 20 Gy (V20Gy) and 40 Gy (V40Gy), though IMRT, VMAT, and PT reduce neurotoxicity [25].
Dosimetric constraints (<54 Gy to brainstem and optic chiasm) limit dose escalation [25]. Delays >6 weeks from surgery to RT worsen OS and PFS, though moderate delays (~6 weeks) may benefit patients with residual disease [33,34].
TTFields extend PFS by 2.7 months (6.7 vs. 4.0 months) but are limited by cost and adherence (≥18 h/day) [35].
Biologically, GBM radioresistance is driven by genetic heterogeneity, including EGFR amplification (57% per TCGA), phosphatase and tensin homolog (PTEN) mutations (40% per TCGA), cyclin-dependent kinase inhibitor 2A/B (CDKN2A/B) deletions (60% per TCGA), TERT promoter mutations (>70% per TCGA), and tumor stem cells [36,39]. Additionally, the immunosuppressive tumor microenvironment, including macrophages/microglia, and neuronal crosstalk promoting proliferation and invasion contribute to resistance.
Poly (ADP-ribose) polymerase (PARP) inhibitors such as veliparib and other radiosensitizers show promise in early-phase trials by inhibiting DNA repair, awaiting phase III confirmation [36,39]. EGFR amplification activates the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathway, conferring RT resistance, with limited benefits from inhibitors like erlotinib [41].
Tumor hypoxia, mediated by hypoxia-inducible factor 1-alpha (HIF-1α), promotes resistance [36]. Strategies like modulated electrohyperthermia (mEHT) and hypoxia-sensitizing agents enhance radiosensitivity, while high-linear energy transfer (LET, energy deposited per unit length) radiation (e.g., carbon ions; alpha particles) is promising but investigational [37]. BNCT generates alpha particles in situ via neutron capture by boron in tumor cells, differing from targeted alpha therapy (TAT) using direct alpha-emitters (e.g., 213Bi, 225Ac, and 211At) conjugated to ligands like substance P for selective tumor targeting (see Section 5.6).
The subventricular zone (SVZ), a reservoir of tumor stem cells with genetic alterations, drives recurrence; irradiating the SVZ with doses ≥56 Gy (ipsilateral) or ≥50 Gy (contralateral) does not significantly improve PFS or OS [42,43].
RT-induced lymphopenia (14% with protons vs. 39% with photons) limits immunotherapy efficacy by depleting CD4 and CD8 T cells, critical for immune checkpoint inhibitors; RT may enhance immunogenicity by inducing tumor antigen release, but the immunosuppressive tumor microenvironment, including regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), restricts benefits (see Section 5.5 for further details) [36,44].
Clinically, pseudoprogression (30–40% with MGMT-methylated) complicates assessment up to 12 weeks [46,47]. Multimodal imaging, including multiparametric MRI and amino acid PET enables RT personalization, achieving a mOS of 23 months in a phase I trial without significant toxicity [48].
Biomarkers (EGFR, PTEN, TERT, and MGMT) enable stratification; only MGMT methylation improves OS in phase III trials [49]. A recent meta-analysis showed that combined targeted therapies improve PFS in newly diagnosed GBM (nGBM) but not OS in nGBM or recurrent GBM (rGBM), reflecting tumor heterogeneity and molecular resistance [50].

4. Molecular Determinants in Glioblastoma Multiforme

Most GBMs, classified as IDH-wt, exhibit aggressive biology and radioresistance due to genetic and epigenetic alterations that enhance DNA repair and cell survival [49]. MGMT methylation (~45%) increases sensitivity to chemoradiation (CRT), while EGFR, PTEN, and TERT contribute to resistance [12,49]. Patients with MGMT methylation, a beneficial prognostic marker, show significantly higher OS (21.7 vs. 15.3 months) and better response to reirradiation in relapse compared to unmethylated cases [12,51,52] (Table 5).
A key radioresistance mechanism in IDH-wt GBMs is activation of the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway, driven by EGFR amplification and PTEN loss, which promotes proliferation, inhibits apoptosis, and enhances DNA repair post-RT [40]. Inhibitors like erlotinib yield modest results (ESMO Scale for Clinical Actionability of Molecular Targets (ESCAT) IIIA) [54]. Other alterations, including phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) mutations and cyclin-dependent kinases 4 and 6 (CDK4/6) amplification, enhance cell cycle progression, while CDKN2A/B homozygous deletion, causing loss of cyclin-dependent kinase inhibitor 2A (p16^INK4a^) and alternate reading frame protein p14 (p14^ARF^), is linked to poor prognosis [49,56]. These biomarkers lack effective targeted therapies due to signaling redundancy and pharmacokinetic limitations in the central nervous system [55]. In contrast, IDH1/2 mutations are absent in IDH-wt GBMs (defining the subtype), but in rare IDH-mutant astrocytoma grade 4 (~10% of grade 4 gliomas), they confer greater radiosensitivity and better prognosis (ESCAT I) [53]. TERT promoter mutations, present in >70% of IDH-wt GBMs, activate telomerase and may promote immune evasion, though their role as predictive markers or therapeutic targets remains unclear [3,57]. Ongoing clinical trials are exploring TERT as a potential immunotherapeutic target, particularly in combination with immune checkpoint inhibitors, to enhance RT efficacy in immunosuppressive tumor microenvironments [3]. Given the limited clinical impact of standard RT in unfavorable molecular subgroups, radiosensitization strategies are being explored to enhance its therapeutic effect [58]. MGMT promoter methylation, a beneficial epigenetic marker, cannot be directly inhibited or induced pharmacologically in clinical practice; MGMT protein inhibitors (e.g., O6-benzylguanine) aim to mimic its effect in unmethylated tumors but are limited by toxicity and are not standard [51].
Among the most promising are poly (ADP-ribose) polymerase (PARP) inhibitors, which interfere with single-strand DNA break repair, exacerbating radiation-induced damage [59]. These inhibitors are particularly relevant for MGMT-unmethylated tumors, where TMZ resistance limits therapeutic options [12]. Although preclinical evidence shows synergy between PARP inhibitors and RT, their clinical efficacy is still under evaluation [49]. A recent randomized phase II trial (VELIPARIB with Radiation Therapy and Temozolomide in Unmethylated MGMT Glioblastoma; VERTU) demonstrated that the PARP inhibitor veliparib, combined with TMZ and RT, improved PFS in MGMT-unmethylated patients (PFS-6m 46% vs. 31% in standard arm), pending phase III confirmation [39]. RT-induced lymphopenia restricts synergy with immunotherapy, but MGMT methylation identifies patients with favorable immune microenvironments, and PT may reduce lymphopenia to enhance combined therapies [44]. Genomic profiling is a promising approach to personalize RT and its combination with targeted therapies or immunotherapy, with EANO 2025 guidelines recommending systematic profiling to optimize diagnosis and identify candidates for personalized trials [58,60].

5. Advances in the Treatment of Glioblastoma Multiforme

GBM is an aggressive brain tumor with a high recurrence rate. In non-elderly patients with good functional status (KPS ≥ 70), the Stupp protocol has been the standard of care for nearly two decades [12]. However, tumor resistance has limited progress over the past decade [36]. Current useful methods emphasize a multimodal strategy: maximal safe resection is followed by RT (60 Gy/30 fractions) with concurrent/adjuvant TMZ as the standard, improving OS especially in MGMT-methylated patients; TTFields are added to maintenance TMZ for survival benefit in selected cases; short-course RT (± TMZ) is leveraged for the elderly/frail; and at recurrence, options like re-resection, reirradiation, or systemic therapies (e.g., lomustine; bevacizumab for symptom control) are used. Investigational approaches like immunotherapies and targeted therapies show promise but lack phase III confirmation, emphasizing the need for randomized controlled trials (RCTs) to validate efficacy.
Integration of advanced medical imaging and nuclear medicine is essential for therapeutic decision-making and personalized GBM management, while supportive and palliative care are crucial throughout.
Below, we discuss promising methods to improve tumor control and quality of life while reducing toxicity, prioritizing RCTs where available.

5.1. Technological Advances in Therapies

Technological advancements have enhanced the precision of treatment, minimizing damage to healthy tissues [61]. PT is increasingly relevant, particularly in reirradiation and selected cases [61]. It leverages the Bragg peak, depositing maximum energy at the end of its range with a sharp fall-off, to target the tumor, with a relative biological effectiveness (RBE) of 1.1 [61]. A phase II RTC (NCT01854554) demonstrated that PT (60 Gy/30 fractions) significantly reduced grade ≥ 3 lymphopenia (14% vs. 39%), minimized irradiated brain volumes (V5–V40), and preserved immune function, potentially enhancing immunotherapy efficacy [6,44]. Although it did not delay cognitive decline compared to IMRT, it reduced fatigue and grade ≥ 2 toxicity, lowering doses to critical structures, suggesting potential for cognitive preservation in low-grade gliomas [6,62]. The NRG-BN001, a phase II RTC (NCT02179086) evaluating dose-escalated photon RT (75 Gy in 30 fractions) versus proton radiotherapy with TMZ, reports improved PFS but no mOS benefit (18.7 months) for the photon arm, with proton arm results pending [63,64]. Accessibility barriers, including high costs and limited centers, restrict PT adoption. Carbon ion radiotherapy (CIRT) achieves a mOS of 18 months in nGBM from retrospective studies [37]. Boron neutron capture therapy (BNCT) reports an mOS of 25.7 months in nGBM with TMZ and 18.9 months in rGBM from non-randomized studies [65,66], with accessibility limited by infrastructure and toxicity concerns (e.g., cerebral edema). Magnetic resonance-guided radiotherapy (MRgRT) achieves an mOS of 18.5 months and a PFS of 11.6 months in nGBM (UNITED, non-randomized phase II), with UNITED2 ongoing [67,68]. TTFields, a standard in NCCN (category 1) and American Society of Clinical Oncology (ASCO) guidelines but not endorsed by the National Institute for Health and Care Excellence (NICE) due to cost-effectiveness, low adherence, and biases in EF-14 (unblinded, selected patients), achieve an mOS of 20.9 months (PFS 6.7 months) in nGBM (phase III EF-14) and an mOS of 10.3 months in rGBM (EF-11). These biases include lack of blinding and selection of healthier patients, potentially inflating efficacy estimates. Trials include phase II 2-THE-TOP (NCT03405792), reporting an mOS of 24.8 months and PFS of 12.0 months with pembrolizumab plus TMZ in nGBM [14], and ongoing phase III trials (TRIDENT with RT/TMZ, EF-41 with TMZ plus pembrolizumab in nGBM) [69,70,71]. Modulated electrohyperthermia (mEHT), laser interstitial thermal therapy (LITT), and magnetic hyperthermia (MHT) have limited evidence in rGBM, supported by non-randomized phase I/II trials [72,73,74,75]. Table 6 details these modalities, emphasizing RCTs where available and noting accessibility barriers. Most of these modalities’ non-randomized or retrospective data limit comparisons with photon-based RT.

5.2. Modified Fractionation Schedules

Modified fractionation schedules optimize RT by counteracting tumor repopulation and reducing treatment duration [36,76]. Hypofractionation, standard in elderly patients with poor functional status (40 Gy/15 fractions), shows promise in younger patients (50–60 Gy/20 fractions with TMZ). The randomized phase II HART-GBM trial achieved an mOS of 26.5 months compared to 22.4 months with standard fractionation though no phase III trials confirm these findings [76]. An institutional study reported an mOS of 19.8 months [77], and a meta-analysis showed a 12-month OS of 71.3% across various ages [78]. Hyperfractionation shows no clear benefit, with similar survival outcomes and moderate toxicity compared to standard fractionation [36,79]. Dose escalation (75 Gy/30 fractions) improves PFS but not mOS (18.7 months) in the NRG-BN001 phase II RTC with TMZ in nGBM; PT arm results pending [63]. Dose escalation with a CIRT boost (16.8–24.8 GyE) achieves an mOS of 18 months [37]. Toxicities include radionecrosis (6.7–14.2%) and cognitive decline [77,80,81]. Tumor heterogeneity and hypoxia necessitate phase III trials, as detailed in Table 7.

5.3. Reirradiation

Reirradiation is a viable option for rGBM in patients with recurrence more than 6 months after initial RT, following multidisciplinary discussion. The 2025 ESTRO/EANO guidelines (KPS > 60; tumor volume < 35 cm3) and ASTRO 2025 guidelines (KPS ≥ 70; tumor volume ≤ 6 cm3) endorse its selective use [11,82]. Post-contrast T1 MRI delineates the GTV, complemented by [18F]-FET or [18F]-FDOPA PET to detect recurrence [82,83]. Regimens such as hypofractionated RT (35 Gy/10 fractions) with bevacizumab (BEV) achieve an mOS of 10.1 months and PFS of 3–6 months, with approximately 5% radionecrosis [82]. NRG Oncology/RTOG 1205 demonstrated improved PFS with hypofractionated RT plus BEV [82]. The LEGATO trial evaluates lomustine with or without reirradiation [84]. Hypofractionated stereotactic RT (25 Gy/5 fractions) showed outcomes similar to 35 Gy/5 fractions, with lower toxicity but higher radionecrosis in larger volumes [85]. CIRT (45 Gy RBE/15 fractions) improves mOS (8.0 months) compared to photons [37]. PT achieves an mOS of 7.8–19.4 months in reirradiation [64]. While not detailed here, brachytherapy, pulsed low-dose-rate radiotherapy (pLDR), and flash radiotherapy are promising for recurrent GBM [11,36]. See Table 8.

5.4. Neoadjuvant Therapy

Neoadjuvant therapy (NAT) reduces tumor volume to facilitate resection or enhance CRT, though it faces challenges such as the need for invasive stereotactic biopsy to confirm diagnosis [86]. In preoperative NAT, BEV in patients with low KPS improves resection (>95%), with an mOS of 15.7 months [87]. POBIG (phase I) evaluates stereotactic RT (6–14 Gy/1 fraction) [88], and PARADIGMA (phase II, NCT03480867) explores RT plus TMZ [86]. In postoperative NAT for unresectable GBM, TMZ plus BEV, the most promising regimen, achieves an mOS of 12.5 months and PFS of 7.4–8.6 months, but BEV increases intracranial hemorrhages [86,89,90]. For resectable GBM, the MAGMA phase III RCT (NCT02394639) evaluates neoadjuvant TMZ (75 mg/m2 daily) and extended adjuvant TMZ (150–200 mg/m2 until progression) before CRT (60 Gy/20 fractions), achieving an mOS of 23 months in MGMT-methylated cases, with hematological toxicity as a primary concern [91].
Neoadjuvant immunotherapies, such as pembrolizumab in resectable rGBM and triple immunotherapy (nivolumab plus ipilimumab plus relatlimab) in nGBM, show potential in selected subgroups [92,93]. Preliminary data from a single nGBM case in the GIANT trial (NCT06816927) suggest no recurrence at 17 months, pending further validation [93]. Future trials should combine immune checkpoint inhibitors (ICIs) with intratumoral oncolytic viruses to enhance immune responses, with strict patient selection due to variable toxicity [86]. ICI immunotherapy combined with oncolytic viruses may benefit selected subgroups [86] (Table 9)

5.5. Immunotherapy, Targeted Therapies, and Chemotherapy

Several immunotherapy trials, notably CheckMate 143, 498, and 548, have failed to demonstrate OS benefits, highlighting challenges in overcoming GBM’s immunosuppressive microenvironment [3,92,94]. Chemotherapies, targeted therapies, and immunotherapies are evaluated for newly diagnosed (nGBM) and recurrent GBM (rGBM) (Table 10).
Standard TMZ chemotherapy, established by the EORTC/NCIC CE.3 trial, is the reference treatment in nGBM, improving survival compared to RT alone [12], though intensive TMZ offers no benefit (RTOG 0525) [95]. In rGBM, metronomic TMZ shows limited activity, particularly after BEV (RESCUE), and lomustine offers modest results (EORTC 26101) [97,98,99]. Lomustine plus TMZ achieves an mOS of 48.1 months in nGBM with MGMT methylation (CeTeG/NOA-09) [96]. Chemotherapy- and RT-induced lymphopenia, as discussed in Section 4, may limit subsequent immunotherapy efficacy.
Targeted therapies, such as BEV, do not extend OS in nGBM (AVAglio, RTOG 0825; mOS 16.8 months; mPFS 10.6 months) but improve disease control in rGBM (BRAIN, EORTC 26101, BELOB; mOS 9.2 months; mPFS 4.2 months) [98,99,100,101] (see Table 9 for its neoadjuvant role). PARP inhibitors, such as veliparib, tested in the VERTU trial, and niraparib (under evaluation in phase 3 trials for MGMT-unmethylated nGBM) are a promising targeted therapy in nGBM with unmethylated MGMT by enhancing radiation-induced damage, one of the few strategies with clinical evidence in specific subgroups [39]. Antibody-drug conjugates (ADCs) targeting EGFR are also emerging in early-phase trials for rGBM. Cilengitide is ineffective in nGBM (CENTRIC, CORE) [99,102], as are erlotinib and everolimus, both inhibitors of tumor signaling pathways [41,99]. Regorafenib has limited efficacy in rGBM (REGOMA), outperforming erlotinib or everolimus but with modest benefits [99]. BRAF/MEK inhibitors show promising responses in BRAF V600E-mutated cases [99]. IDH inhibitors, such as vorasidenib, are under investigation for IDH-mutant gliomas, with no data in GBM [99].
In immunotherapy, nivolumab does not outperform BEV in rGBM (CheckMate 143) or TMZ with RT in nGBM with unmethylated (CheckMate 498) or MGMT-methylated (CheckMate 548), where treatment-induced lymphopenia may reduce efficacy [3,92,94]. Adoptive cellular therapies, such as CAR-T, and oncolytic viruses, such as G47Δ (modified herpes simplex virus) or DNX-2401 (adenovirus), are promising in rGBM. DNX-2401, in monotherapy (20% 3-year survival) or combined with pembrolizumab in the CAPTIVE trial (mOS 12.5 months, ongoing), shows preliminary efficacy, though immunosuppression from dexamethasone and RT may limit its effectiveness [92,105]. Neoadjuvant pembrolizumab before surgery in rGBM improves survival with an mOS of 13.8 months in specific subgroups [92,104] (see Table 9 for neoadjuvant use). The DCVax-L vaccine benefits nGBM (mOS 19.3 months) and rGBM (mOS 13.2 months) but is not standard; rindopepimut does not improve survival in EGFRvIII-positive nGBM (ACT IV) [92,106]. Other vaccines are in development for GBM [92]. Cytokines like interferon-alpha (IFN-α) enhance TMZ in nGBM (mOS 26.7 months, phase III), though not standard, and neoadjuvant triple immunotherapy before surgery prevents recurrences in isolated nGBM cases (GIANT, ongoing) [3,93]. The blood–brain barrier (BBB) and treatment-induced immunosuppression limit the combination of immunotherapy with RT [92] (Table 10)

5.6. Advanced Imaging and Theranostics

Multiparametric MRI, including T1/T2-FLAIR, dynamic susceptibility contrast perfusion (DSC), diffusion-weighted imaging (DWI), and magnetic resonance spectroscopy (MRS), detects gliomas with high sensitivity, evaluates recurrence per RANO 2.0 criteria, including non-enhancing disease in IDH-mutant gliomas, and distinguishes pseudoprogression from true progression using DSC, DWI, and MRS (Table 11) [47,107,108], further enhancing early GBM diagnosis and personalized RT planning by detecting non-enhancing tumors with MRI/PET and optimizing tumor control probability using radiobiological models like Poisson to tailor dose prescriptions [48,109].
Challenges include high costs and the need for specialized expertise in interpreting multiparametric MRI [107,108]. PET with amino acid tracers ([18F]-FET, [11C]-MET, [18F]-FDOPA, [18F]-FACBC) and [68Ga]-PSMA-11 differentiates recurrence from pseudoprogression with high specificity [110,111,112,113]. These techniques optimize RT planning by improving tumor delineation and enabling dose escalation in high-risk regions. A phase I trial using multiparametric MRI and [18F]-FDOPA PET achieved an mOS of 23 months without significant toxicity, though phase III trials have not confirmed OS benefits [48]. Theranostics with [131I]-IPA achieves an mOS of 16 months in rGBM, limited by BBB penetration [111] (Table 11). Ongoing trials are evaluating novel theranostic agents [111,113]. Alpha particle-emitting radiopharmaceuticals, delivering high-LET radiation via ligands such as substance P targeting hypoxic tumor cells, show promise in preclinical and early clinical studies for GBM with minimal damage to surrounding healthy tissue due to their short range and high cytotoxicity but remain investigational [38].

5.7. Artificial Intelligence in GBM Radiotherapy

Artificial Intelligence (AI) revolutionizes GBM RT by integrating multiparametric MRI and PET data to enhance treatment planning, predict recurrence, and personalize therapy through advanced radiomic analysis [48,109,114]. AI-driven algorithms automate tumor segmentation, quantify biological heterogeneity, and differentiate true progression from pseudoprogression, improving target volume delineation and adaptive dose strategies [48,114]. By leveraging radiobiological models like Poisson, AI optimizes Tumor Control Probability (TCP) calculations, tailoring doses to tumor characteristics and biomarkers such as MGMT methylation [109,114]. Challenges include data variability, non-standardized imaging protocols, and limited model interpretability, necessitating multicenter prospective trials to validate clinical efficacy and reproducibility of AI-driven TCP optimization [48,114].

6. Conclusions

Despite significant advancements in GBM management, RT continues to face inherent limitations, primarily due to diffuse tumor infiltration and intrinsic radioresistance, often driven by specific molecular alterations like EGFR amplification, MGMT methylation, and TERT promoter mutations, alongside challenges from the immunosuppressive microenvironment and cancer neuroscience.
Advanced RT techniques, including IMRT, PT, CIRT, BNCT, TTFields, and localized therapies like MHT and LITT for rGBM, optimize dose precision, yet recurrences remain common.
Promising strategies such as neoadjuvant approaches, modified fractionation like hypofractionated RT, and reirradiation for recurrent GBM show therapeutic potential.
The integration of systemic therapies, including immunotherapies, chemotherapies, and targeted agents, is crucial for enhancing tumor control, though RT-induced lymphopenia limits immunotherapy efficacy. Multiparametric MRI and PET enable tailored RT planning, while radiotheranostics like [131I]-IPA broaden treatment options, despite BBB challenges. To advance GBM management, phase III RCTs are needed to validate dose-escalation strategies with PT, biomarker-driven trials to personalize therapies, and AI integration for adaptive RT planning to differentiate true progression from pseudoprogression.

7. Future Directions

The future of GBM radiotherapy lies in leveraging molecular biomarkers and advanced imaging (e.g., multiparametric MRI; amino acid PET) to enable tailored treatment planning. Biomarker-driven clinical trials are critical to personalize RT and systemic therapies, integrating molecular profiles to overcome radioresistance. Phase III RCTs are urgently needed to validate PT dose escalation, with the phase II NRG-BN001 trial (NCT02179086) designed to assess a threshold for advancing to phase III, though preliminary photon arm results did not meet criteria, and proton arm results are pending. Other phase III RCTs, such as TRIDENT (NCT04471844) and EF-41 (NCT06556363) for TTFields with systemic therapies, and LEGATO (NCT04078568) for reirradiation, aim to confirm clinical benefits. Emerging trials, including the phase 3 Gliofocus (NCT06388733) comparing niraparib vs. TMZ in newly diagnosed MGMT-unmethylated GBM, may address TMZ challenges in resistant subgroups [103]. According to the 2025 SNO-EANO consensus, strategies to overcome the BBB, such as focused ultrasound or local delivery, and liquid biopsy technologies to monitor tumor evolution are promising to enhance therapeutic delivery and personalization [45].
AI-driven predictive models can achieve precise, adaptive RT planning by integrating tumor biology and imaging data, enhancing differentiation of true progression from pseudoprogression and optimizing dose delivery. Ethical considerations, such as mitigating biases in AI training data, are crucial for clinical adoption.
Radiotheranostics, including [131I]-IPA and agents like [177Lu]-PSMA, and alpha particle-emitting radiopharmaceuticals for targeted therapy with high LET to address hypoxia, hold promise for targeted imaging and therapy, improving oncological outcomes and quality of life by overcoming blood–brain barrier challenges, with phase I/II trials showing improved PFS and potential synergy with RT to elevate median mOS in rGBM.

Author Contributions

Conceptualization, C.F.; methodology, C.F.; literature review, C.F., R.C., A.D. and I.G.; writing—original draft preparation, C.F., R.C., A.D. and I.G.; writing—review and editing, C.F., R.C., A.D. and I.G.; data curation, R.C.; supervision, C.F. and F.C.; resources, F.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article as all data discussed are derived from publicly available sources cited in the references.

Conflicts of Interest

F.C. declares the following conflicts of interest: sponsored research support from Janssen; consulting or advisory roles with Janssen and AstraZeneca; honoraria for lectures or educational activities from Janssen, Roche Farma, AstraZeneca, Astellas, Recordati, and Ipsen; and travel, accommodation, or expense support from AstraZeneca, Astellas, Janssen, and Roche Farma. The remaining authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
3D-CRTThree-Dimensional Conformal Radiotherapy
ADCApparent Diffusion Coefficient
ADCsAntibody-Drug Conjugates
AIArtificial Intelligence
AMFAlternating Magnetic Field
ASCOAmerican Society of Clinical Oncology
ASTROAmerican Society for Radiation Oncology
BBBBlood–Brain Barrier
BEVBevacizumab
BNCTBoron Neutron Capture Therapy
BSHSodium Borocaptate
CDK4/6Cyclin-Dependent Kinases 4 and 6
CDKN2A/BCyclin-Dependent Kinase Inhibitor 2A/B
CFRTConventional Fractionated Radiotherapy
ChoCholine
CIRTCarbon Ion Radiotherapy
CrCreatine
CRTChemoradiation
CTChemotherapy
CTVClinical Target Volume
ddTMZDense Dose Temozolomide
DSCDynamic Susceptibility Contrast Perfusion
DWIDiffusion-Weighted Imaging
EGFREpidermal Growth Factor Receptor
EORTCEuropean Organisation for Research and Treatment of Cancer
ESCATESMO Scale for Clinical Actionability of Molecular Targets
ESTROEuropean Society for Radiotherapy and Oncology
EANOEuropean Association of Neuro-Oncology
FSRTFractionated Stereotactic Radiotherapy
GBMGlioblastoma Multiforme
GTVGross Tumor Volume
GyEGray Equivalent
HFRTHypofractionated Radiotherapy
HIF-1αHypoxia-Inducible Factor 1-Alpha
HRHazard Ratio
HSRTHypofractionated Stereotactic Radiotherapy
ICIImmune Checkpoint Inhibitor
IDHIsocitrate Dehydrogenase
IGRTImage-Guided Radiotherapy
IMPTIntensity-Modulated Proton Therapy
IMRTIntensity-Modulated Radiotherapy
MDSCsMyeloid-Derived Suppressor Cells
KPSKarnofsky Performance Status
L-BPAL-4-Boronophenylalanine
LITTLaser Interstitial Thermal Therapy
MDACCMD Anderson Cancer Center
mEHTModulated Electrohyperthermia
MHTMagnetic Hyperthermia
MNPMagnetic Nanoparticles
mOSMedian Overall Survival
MRIMagnetic Resonance Imaging
MRSMagnetic Resonance Spectroscopy
MRgRTMagnetic Resonance-Guided Radiotherapy
NAAN-Acetyl Aspartate
NATNeoadjuvant Therapy
NCCNNational Comprehensive Cancer Network
NICENational Institute for Health and Care Excellence
nGBMNewly Diagnosed Glioblastoma Multiforme
OSOverall Survival
PARPPoly (ADP-Ribose) Polymerase
PETPositron Emission Tomography
PFSProgression-Free Survival
PI3K/AKTPhosphatidylinositol 3-Kinase/Protein Kinase B
PIK3CAPhosphatidylinositol-4,5-Bisphosphate 3-Kinase Catalytic Subunit Alpha
PTProton Therapy
PTENPhosphatase and Tensin Homolog
PTVPlanning Target Volume
RANOResponse Assessment in Neuro-Oncology
RBERelative Biological Effectiveness
rGBMRecurrent Glioblastoma Multiforme
RTRadiotherapy
RCTsRandomized Controlled Trials
SRSStereotactic Radiosurgery
SVZSubventricular Zone
TATTargeted Alpha Therapy
TCGAThe Cancer Genome Atlas
TCPTumor Control Probability
TERTTelomerase Reverse Transcriptase
TMZTemozolomide
TP53Tumor Protein p53
TTFieldsTumor Treating Fields
V20GyVolume Receiving 20 Gray
V40GyVolume Receiving 40 Gray
VMATVolumetric Modulated Arc Therapy
WBRTWhole-Brain Radiotherapy
WHOWorld Health Organization
wtWild-Type

References

  1. Ostrom, Q.T.; Gittleman, H.; Xu, J.; Kromer, C.; Wolinsky, Y.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the United States in 2009–2013. Neuro-Oncology 2016, 18 (Suppl. 5), v1–v75. [Google Scholar] [CrossRef] [PubMed]
  2. Segura, P.P.; Quintela, N.V.; García, M.M.; del Barco Berrón, S.; Sarrió, R.G.; Gómez, J.G.; Castaño, A.G.; Martín, L.M.N.; Rubio, O.G.; Losada, E.P. SEOM-GEINO clinical guidelines for high-grade gliomas of adulthood (2022). Clin. Transl. Oncol. 2023, 25, 2634–2646. [Google Scholar] [CrossRef]
  3. Schaff, L.R.; Mellinghoff, I.K. A review of glioblastoma and other primary brain malignancies—Reply. JAMA 2023, 330, 189–190. [Google Scholar] [CrossRef] [PubMed]
  4. Wanis, H.A.; Møller, H.; Ashkan, K.; Davies, E.A. The incidence of major subtypes of primary brain tumors in adults in England 1995–2017. Neuro-Oncology 2021, 23, 1371–1382. [Google Scholar] [CrossRef]
  5. Louis, D.N.; Perry, A.; Wesseling, P.; Brat, D.J.; Cree, I.A.; Figarella-Branger, D.; Hawkins, C.; Ng, H.K.; Pfister, S.M.; Reifenberger, G.; et al. The 2021 WHO classification of tumors of the central nervous system: A summary. Neuro-Oncology 2021, 23, 1231–1251. [Google Scholar] [CrossRef] [PubMed]
  6. Brown, N.F.; Ottaviani, D.; Tazare, J.; Gregson, J.; Kitchen, N.; Brandner, S.; Fersht, N.; Mulholland, P. Survival outcomes and prognostic factors in glioblastoma. Cancers 2022, 14, 3161. [Google Scholar] [CrossRef]
  7. Gorlia, T.; van den Bent, M.J.; Hegi, M.E.; Mirimanoff, R.O.; Weller, M.; Cairncross, J.G.; Eisenhauer, E.; Belanger, K.; Brandes, A.A.; Allgeier, A.; et al. Nomograms for predicting survival of patients with newly diagnosed glioblastoma: Prognostic factor analysis of EORTC and NCIC trial 26981-22981/CE.3. Lancet Oncol. 2008, 9, 29–38. [Google Scholar] [CrossRef]
  8. Sipos, D.; Raposa, B.L.; Freihat, O.; Simon, M.; Mekis, N.; Cornacchione, P.; Kovács, Á. Glioblastoma: Clinical presentation, multidisciplinary management, and long-term outcomes. Cancers 2025, 17, 146. [Google Scholar] [CrossRef]
  9. Aboubakr, O.; Moiraghi, A.; Elia, A.; Tauziede-Espariat, A.; Roux, A.; Leclerc, A.; Planet, M.; Bedioui, A.; Simboli, G.A.; Dhermain, F.; et al. Long-term survivors in 976 supratentorial glioblastoma, IDH-wildtype patients. J. Neurosurg. 2025, 142, 174–186. [Google Scholar] [CrossRef]
  10. Mason, W.P.; Harrison, R.A.; Lapointe, S.; Lim-Fat, M.J.; MacNeil, M.V.; Mathieu, D.; Perry, J.R.; Pitz, M.W.; Roberge, D.; Tsang, D.S.; et al. Canadian expert consensus recommendations for the diagnosis and management of glioblastoma: Results of a Delphi study. Curr. Oncol. 2025, 32, 207–220. [Google Scholar] [CrossRef]
  11. Yeboa, D.N.; Braunstein, S.E.; Cabrera, A.; Crago, K.; Galanis, E.; Hattab, E.M.; Heron, D.E.; Huang, J.; Kim, M.M.; Kirkpatrick, J.P.; et al. Radiation therapy for WHO grade 4 adult-type diffuse glioma: An ASTRO clinical practice guideline. Pract. Radiat. Oncol. 2025, 15, 45–67. [Google Scholar] [CrossRef]
  12. Stupp, R.; Mason, W.P.; van den Bent, M.J.; Weller, M.; Fisher, B.; Taphoorn, M.J.B.; Belanger, K.; Brandes, A.A.; Marosi, C.; Bogdahn, U.; et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 2005, 352, 987–996. [Google Scholar] [CrossRef]
  13. Sulman, E.P.; Ismaila, N.; Armstrong, T.S.; Tsien, C.; Batchelor, T.T.; Cloughesy, T.; Galanis, E.; Gilbert, M.; Gondi, V.; Lovely, M.; et al. Radiation therapy for glioblastoma: American Society of Clinical Oncology clinical practice guideline endorsement of the American Society for Radiation Oncology guideline. J. Clin. Oncol. 2017, 35, 361–369. [Google Scholar] [CrossRef] [PubMed]
  14. Cabrera, A.R.; Kirkpatrick, J.P.; Fiveash, J.B.; Shih, H.A.; Koay, E.J.; Lutz, S.; Petit, J.; Chao, S.T.; Brown, P.D.; Vogelbaum, M.; et al. Radiation therapy for glioblastoma: Executive summary of an American Society for Radiation Oncology evidence-based clinical practice guideline. Pract. Radiat. Oncol. 2016, 6, 217–225. [Google Scholar] [CrossRef] [PubMed]
  15. Balana, C.; Vaz, M.A.; Manuel Sepúlveda, J.; Mesia, C.; Del Barco, S.; Pineda, E.; Muñoz-Langa, J.; Estival, A.; de Las Peñas, R.; Fuster, J.; et al. A phase II randomized, multicenter, open-label trial of continuing adjuvant temozolomide beyond six cycles in patients with glioblastoma (GEINO 14-01). Neuro-Oncology 2020, 22, 1851–1861. [Google Scholar] [CrossRef] [PubMed]
  16. Stupp, R.; Taillibert, S.; Kanner, A.A.; Kesari, S.; Steinberg, D.M.; Toms, S.A.; Taylor, L.P.; Lieberman, F.; Silvani, A.; Fink, K.L.; et al. Maintenance therapy with tumor-treating fields plus temozolomide vs temozolomide alone for glioblastoma: A randomized clinical trial. JAMA 2015, 314, 2535–2543. [Google Scholar] [CrossRef]
  17. Roubil, J.; Harris, T. Radiotherapeutic approaches in the treatment of adult gliomas. Adv. Cancer Res. 2025, 163, 153–182. [Google Scholar] [CrossRef]
  18. Perry, J.R.; Laperriere, N.; O’Callaghan, C.J.; Brandes, A.A.; Menten, J.; Phillips, C.; Fay, M.; Nishikawa, R.; Cairncross, J.G.; Roa, W.; et al. Short-course radiation plus temozolomide in elderly patients with glioblastoma. N. Engl. J. Med. 2017, 376, 1027–1037. [Google Scholar] [CrossRef]
  19. Roa, W.; Brasher, P.M.A.; Bauman, G.; Anthes, M.; Bruera, E.; Chan, A.; Fisher, B.; Fulton, D.; Gulavita, S.; Hao, C.; et al. Abbreviated course of radiation therapy in older patients with glioblastoma multiforme: A prospective randomized clinical trial. J. Clin. Oncol. 2004, 22, 1583–1588. [Google Scholar] [CrossRef]
  20. Roa, W.; Kepka, L.; Kumar, N.; Sinaika, V.; Matiello, J.; Lomidze, D.; Hentati, D.; Guedes de Castro, D.; Dyttus-Cebulok, K.; Drodge, S.; et al. International Atomic Energy Agency randomized phase III study of radiation therapy in elderly and/or frail patients with newly diagnosed glioblastoma multiforme. J. Clin. Oncol. 2015, 33, 4145–4150. [Google Scholar] [CrossRef]
  21. Minniti, G.; De Sanctis, V.; Muni, R.; Rasio, D.; Lanzetta, G.; Bozzao, A.; Osti, M.F.; Salvati, M.; Valeriani, M.; Cantore, G.P.; et al. Hypofractionated radiotherapy followed by adjuvant chemotherapy with temozolomide in elderly patients with glioblastoma. J. Neurooncol. 2009, 91, 95–100. [Google Scholar] [CrossRef] [PubMed]
  22. Malmström, A.; Grønberg, B.H.; Marosi, C.; Stupp, R.; Frappaz, D.; Schultz, H.; Abacioglu, U.; Tavelin, B.; Lhermitte, B.; Hegi, M.E.; et al. Temozolomide versus standard 6-week radiotherapy versus hypofractionated radiotherapy in patients older than 60 years with glioblastoma: The Nordic randomised, phase 3 trial. Lancet Oncol. 2012, 13, 916–926. [Google Scholar] [CrossRef] [PubMed]
  23. de Melo, S.M.; Marta, G.N.; de Latorraca, C.O.C.; Martins, C.B.; Efthimiou, O.; Riera, R. Hypofractionated radiotherapy for newly diagnosed elderly glioblastoma patients: A systematic review and network meta-analysis. PLoS ONE 2021, 16, e0257384. [Google Scholar] [CrossRef]
  24. Hanna, C.; Lawrie, T.A.; Rogozińska, E.; Kernohan, A.; Jefferies, S.; Bulbeck, H.; Ali, U.M.; Robinson, T.; Grant, R. Treatment of newly diagnosed glioblastoma in the elderly: A network meta-analysis. Cochrane Database Syst. Rev. 2020, 3, CD013261. [Google Scholar] [CrossRef]
  25. Niyazi, M.; Andratschke, N.; Bendszus, M.; Chalmers, A.J.; Erridge, S.C.; Galldiks, N.; Lagerwaard, F.J.; Navarria, P.; Munck Af Rosenschöld, P.; Ricardi, U.; et al. ESTRO-EANO guideline on target delineation and radiotherapy details for glioblastoma. Radiother. Oncol. 2023, 184, 109663. [Google Scholar] [CrossRef]
  26. Niyazi, M.; Brada, M.; Chalmers, A.J.; Combs, S.E.; Erridge, S.C.; Fiorentino, A.; Grosu, A.L.; Lagerwaard, F.J.; Minniti, G.; Mirimanoff, R.-O.; et al. ESTRO-ACROP Guideline “Target Delineation of Glioblastomas”. Radiother. Oncol. J. Eur. Soc. Ther. Radiol. Oncol. 2016, 118, 35–42. [Google Scholar] [CrossRef]
  27. Kumar, N.; Kumar, R.; Sharma, S.C.; Mukherjee, A.; Khandelwal, N.; Tripathi, M.; Miriyala, R.; Oinam, A.S.; Madan, R.; Yadav, B.S.; et al. Impact of Volume of Irradiation on Survival and Quality of Life in Glioblastoma: A Prospective, Phase 2, Randomized Comparison of RTOG and MDACC Protocols. Neuro-Oncology. Pract. 2020, 7, 86–93. [Google Scholar] [CrossRef]
  28. Minniti, G.; Amelio, D.; Amichetti, M.; Salvati, M.; Muni, R.; Bozzao, A.; Lanzetta, G.; Scarpino, S.; Arcella, A.; Enrici, R.M. Patterns of Failure and Comparison of Different Target Volume Delineations in Patients with Glioblastoma Treated with Conformal Radiotherapy plus Concomitant and Adjuvant Temozolomide. Radiother. Oncol. J. Eur. Soc. Ther. Radiol. Oncol. 2010, 97, 377–381. [Google Scholar] [CrossRef]
  29. Chang, E.L.; Akyurek, S.; Avalos, T.; Rebueno, N.; Spicer, C.; Garcia, J.; Famiglietti, R.; Allen, P.K.; Chao, K.S.C.; Mahajan, A.; et al. Evaluation of Peritumoral Edema in the Delineation of Radiotherapy Clinical Target Volumes for Glioblastoma. Int. J. Radiat. Oncol. Biol. Phys. 2007, 68, 144–150. [Google Scholar] [CrossRef]
  30. Karp, J.M.; Kruser, T.J. Contouring with FLAIR: Targeting Peritumoral Edema (and Beyond) in Glioblastoma. Int. J. Radiat. Oncol. Biol. Phys. 2025, 121, 1182–1184. [Google Scholar] [CrossRef]
  31. Qiu, Y.; Li, Y.; Jiang, C.; Wu, X.; Liu, W.; Fan, C.; Ye, X.; He, L.; Xiao, S.; Zhao, Q.; et al. Toxicity and Efficacy of Different Target Volume Delineations of Radiation Therapy Based on the Updated Radiation Therapy Oncology Group/National Research Group and European Organization for Research and Treatment of Cancer Guidelines in Patients with Glioma. Int. J. Radiat. Oncol. Biol. Phys. 2025, 121, 1168–1181. [Google Scholar] [CrossRef]
  32. Wallner, K.E.; Galicich, J.H.; Krol, G.; Arbit, E.; Malkin, M.G. Patterns of Failure Following Treatment for Glioblastoma Multiforme and Anaplastic Astrocytoma. Int. J. Radiat. Oncol. Biol. Phys. 1989, 16, 1405–1409. [Google Scholar] [CrossRef] [PubMed]
  33. Magrowski, Ł.; Nowicka, E.; Masri, O.; Tukiendorf, A.; Tarnawski, R.; Miszczyk, M. The Survival Impact of Significant Delays between Surgery and Radiochemotherapy in Glioblastoma Patients: A Retrospective Analysis from a Large Tertiary Center. J. Clin. Neurosci. 2021, 90, 39–47. [Google Scholar] [CrossRef] [PubMed]
  34. Zhang, M.; Xu, F.; Ni, W.; Qi, W.; Cao, W.; Xu, C.; Chen, J.; Gao, Y. Survival Impact of Delaying Postoperative Chemoradiotherapy in Newly Diagnosed Glioblastoma Patients. Transl. Cancer Res. 2020, 9, 5450–5458. [Google Scholar] [CrossRef] [PubMed]
  35. Stupp, R.; Taillibert, S.; Kanner, A.; Read, W.; Steinberg, D.; Lhermitte, B.; Toms, S.; Idbaih, A.; Ahluwalia, M.S.; Fink, K.; et al. Effect of Tumor-Treating Fields Plus Maintenance Temozolomide vs Maintenance Temozolomide Alone on Survival in Patients with Glioblastoma: A Randomized Clinical Trial. JAMA 2017, 318, 2306–2316. [Google Scholar] [CrossRef]
  36. Frosina, G. Radiotherapy of High-Grade Gliomas: Dealing with a Stalemate. Crit. Rev. Oncol. Hematol. 2023, 190, 104110. [Google Scholar] [CrossRef]
  37. Koosha, F.; Ahmadikamalabadi, M.; Mohammadi, M. Review of recent improvements in carbon ion radiation therapy in the treatment of glioblastoma. Adv. Radiat. Oncol. 2024, 9, 101465. [Google Scholar] [CrossRef]
  38. Kunikowska, J.; Morgenstern, A.; Pełka, K.; Bruchertseifer, F.; Królicki, L. Targeted alpha therapy for glioblastoma. Front. Med. 2022, 9, 1085245. [Google Scholar] [CrossRef]
  39. Sim, H.W.; McDonald, K.L.; Lwin, Z.; Barnes, E.H.; Rosenthal, M.; Foote, M.C.; Koh, E.S.; Back, M.; Wheeler, H.; Sulman, E.P.; et al. A Randomized Phase II Trial of Veliparib, Radiotherapy, and Temozolomide in Patients with Unmethylated MGMT Glioblastoma: The VERTU Study. Neuro-Oncology 2021, 23, 1736–1749. [Google Scholar] [CrossRef]
  40. Brennan, C.W.; Verhaak, R.G.; McKenna, A.; Campos, B.; Noushmehr, H.; Salama, S.R.; Zheng, S.; Chakravarty, D.; Sanborn, J.Z.; Berman, S.H.; et al. The Somatic Genomic Landscape of Glioblastoma. Cell 2013, 155, 462–477. [Google Scholar] [CrossRef]
  41. van den Bent, M.J.; Brandes, A.A.; Rampling, R.; Kouwenhoven, M.C.; Kros, J.M.; Carpentier, A.F.; Clement, P.M.; Frenay, M.; Campone, M.; Baurain, J.F.; et al. Randomized Phase II Trial of Erlotinib versus Temozolomide or Carmustine in Recurrent Glioblastoma: EORTC Brain Tumor Group Study 26034. J. Clin. Oncol. 2009, 27, 1268–1274. [Google Scholar] [CrossRef] [PubMed]
  42. Matarredona, E.R.; Pastor, A.M. Neural Stem Cells of the Subventricular Zone as the Origin of Human Glioblastoma Stem Cells: Therapeutic Implications. Front. Oncol. 2019, 9, 779. [Google Scholar] [CrossRef] [PubMed]
  43. Mathew, B.S.; Kaliyath, S.B.; Krishnan, J.; Bhasi, S. Impact of Subventricular Zone Irradiation on Outcome of Patients with Glioblastoma. J. Cancer Res. Ther. 2018, 14, 1202–1206. [Google Scholar] [CrossRef] [PubMed]
  44. Mohan, R.; Liu, A.Y.; Brown, P.D.; Mahajan, A.; Dinh, J.; Chung, C.; McAvoy, S.; McAleer, M.F.; Lin, S.H.; Li, J.; et al. Proton Therapy Reduces the Likelihood of High-Grade Radiation-Induced Lymphopenia in Glioblastoma Patients: Phase II Randomized Study of Protons vs Photons. Neuro-Oncology 2021, 23, 284–294. [Google Scholar] [CrossRef]
  45. Wen, P.Y.; Weller, M.; Lee, E.Q.; Touat, M.; Khasraw, M.; Rahman, R.; Platten, M.; Lim, M.; Winkler, F.; Horbinski, C.; et al. Glioblastoma in Adults: A Society for Neuro-Oncology (SNO) and European Society of Neuro-Oncology (EANO) Consensus Review on Current Management and Future Directions. Neuro Oncol. 2025, 27, 1411–1427. [Google Scholar] [CrossRef]
  46. Brandes, A.A.; Franceschi, E.; Tosoni, A.; Blatt, V.; Pession, A.; Tallini, G.; Bertorelle, R.; Bartolini, S.; Calbucci, F.; Andreoli, A.; et al. MGMT Promoter Methylation Status Can Predict the Incidence and Outcome of Pseudoprogression after Concomitant Radiochemotherapy in Newly Diagnosed Glioblastoma Patients. J. Clin. Oncol. 2008, 26, 2192–2197. [Google Scholar] [CrossRef]
  47. Wen, P.Y.; van den Bent, M.; Youssef, G.; Cloughesy, T.F.; Ellingson, B.M.; Weller, M.; Galanis, E.; Barboriak, D.P.; de Groot, J.; Gilbert, M.R.; et al. RANO 2.0: Update to the Response Assessment in Neuro-Oncology Criteria for High- and Low-Grade Gliomas in Adults. J. Clin. Oncol. 2023, 41, 5187–5199. [Google Scholar] [CrossRef]
  48. Breen, W.G.; Aryal, M.P.; Cao, Y.; Kim, M.M. Integrating Multi-Modal Imaging in Radiation Treatments for Glioblastoma. Neuro Oncol. 2024, 26 (Suppl. 2), S17–S25. [Google Scholar] [CrossRef]
  49. Tan, A.C.; Ashley, D.M.; López, G.Y.; Malinzak, M.; Friedman, H.S.; Khasraw, M. Management of Glioblastoma: State of the Art and Future Directions. CA Cancer J. Clin. 2020, 70, 299–312. [Google Scholar] [CrossRef]
  50. Ma, Y.; Wang, Y.; Nie, C.; Lin, Y. The Efficacy of Targeted Therapy Combined with Radiotherapy and Temozolomide-Based Chemotherapy in the Treatment of Glioma: A Systemic Review and Meta-Analysis of Phase II/III Randomized Controlled Trials. Front. Oncol. 2023, 13, 1082539. [Google Scholar] [CrossRef]
  51. Hegi, M.E.; Diserens, A.C.; Gorlia, T.; Hamou, M.F.; de Tribolet, N.; Weller, M.; Kros, J.M.; Hainfellner, J.A.; Mason, W.; Mariani, L.; et al. MGMT Gene Silencing and Benefit from Temozolomide in Glioblastoma. N. Engl. J. Med. 2005, 352, 997–1003. [Google Scholar] [CrossRef]
  52. Weller, M.; Stupp, R.; Reifenberger, G.; Brandes, A.A.; van den Bent, M.J.; Wick, W.; Hegi, M.E. MGMT Promoter Methylation in Malignant Gliomas: Ready for Personalized Medicine? Nat. Rev. Neurol. 2010, 6, 39–51. [Google Scholar] [CrossRef]
  53. Yan, H.; Parsons, D.W.; Jin, G.; McLendon, R.; Rasheed, B.A.; Yuan, W.; Kos, I.; Batinic-Haberle, I.; Jones, S.; Riggins, G.J.; et al. IDH1 and IDH2 mutations in gliomas. N. Engl. J. Med. 2009, 360, 765–773. [Google Scholar] [CrossRef] [PubMed]
  54. Brown, P.D.; Krishnan, S.; Sarkaria, J.N.; Wu, W.; Jaeckle, K.A.; Uhm, J.H.; Geoffroy, F.J.; Arusell, R.; Kitange, G.; Jenkins, R.B.; et al. Phase I/II Trial of Erlotinib and Temozolomide with Radiation Therapy in the Treatment of Newly Diagnosed Glioblastoma Multiforme: North Central Cancer Treatment Group Study N0177. J. Clin. Oncol. 2008, 26, 5603–5609. [Google Scholar] [CrossRef] [PubMed]
  55. Wen, P.Y.; Weller, M.; Lee, E.Q.; Alexander, B.M.; Barnholtz-Sloan, J.S.; Barthel, F.P.; Batchelor, T.T.; Bindra, R.S.; Chang, S.M.; Chiocca, E.A.; et al. Glioblastoma in adults: A Society for Neuro-Oncology (SNO) and European Society of Neuro-Oncology (EANO) consensus review on current management and future directions. Neuro Oncol. 2020, 22, 1073–1113. [Google Scholar] [CrossRef] [PubMed]
  56. Ma, S.; Rudra, S.; Campian, J.L.; Dahiya, S.; Dunn, G.P.; Johanns, T.; Goldstein, M.; Kim, A.H.; Huang, J. Prognostic impact of CDKN2A/B deletion, TERT mutation, and EGFR amplification on histological and molecular IDH-wildtype glioblastoma. Neuro-Oncol. Adv. 2020, 2, vdaa126. [Google Scholar] [CrossRef]
  57. Barthel, F.P.; Johnson, K.C.; Varn, F.S.; Moskalik, A.D.; Tanner, G.; Kocakavuk, E.; Anderson, K.J.; Abiola, O.; Aldape, K.; Alfaro, K.D.; et al. Longitudinal molecular trajectories of diffuse glioma in adults. Nature 2019, 576, 112–120. [Google Scholar] [CrossRef]
  58. van den Bent, M.J.; Franceschi, E.; Touat, M.; French, P.J.; Idbaih, A.; Lombardi, G.; Rudà, R.; Schweizer, L.; Capper, D.; Sanson, M.; et al. Updated EANO guideline on rational molecular testing of gliomas, glioneuronal, and neuronal tumors in adults for targeted therapy selection-update 1. Neuro Oncol. 2025, 27, 331–337. [Google Scholar] [CrossRef]
  59. Dungey, F.A.; Löser, D.A.; Chalmers, A.J. Replication-dependent radiosensitization of human glioma cells by inhibition of poly(ADP-ribose) polymerase: Mechanisms and therapeutic potential. Int. J. Radiat. Oncol. Biol. Phys. 2008, 72, 1188–1197. [Google Scholar] [CrossRef]
  60. Kotecha, R.; Odia, Y.; Khosla, A.A.; Ahluwalia, M.S. Key clinical principles in the management of glioblastoma. JCO Oncol. Pract. 2023, 19, 180–189. [Google Scholar] [CrossRef]
  61. Corrales-García, E.M.; Aristu-Mendioroz, J.J.; Castro-Novais, J.; Matute-Martín, R.; Learra-Martínez, M.C.; Delgado-López, P.D. Current state of proton therapy for tumors of the central nervous system in Spain: Physical bases, indications, controversies and perspectives. Clin. Transl. Oncol. 2025, 27, 858–870. [Google Scholar] [CrossRef]
  62. Brown, P.D.; Chung, C.; Liu, D.D.; McAvoy, S.; Grosshans, D.; Al Feghali, K.; Mahajan, A.; Li, J.; McGovern, S.L.; McAleer, M.F.; et al. A prospective phase II randomized trial of proton radiotherapy vs intensity-modulated radiotherapy for patients with newly diagnosed glioblastoma. Neuro Oncol. 2021, 23, 1337–1347. [Google Scholar] [CrossRef] [PubMed]
  63. Gondi, V.; Pugh, S.; Tsien, C.; Chenevert, T.; Gilbert, M.; Omuro, A.; Mcdonough, J.; Aldape, K.; Srinivasan, A.; Rogers, C.L.; et al. Radiotherapy (RT) dose-intensification (DI) using intensity-modulated RT (IMRT) versus standard-dose (SD) RT with temozolomide (TMZ) in newly diagnosed glioblastoma (GBM): Preliminary results of NRG Oncology BN001. Int. J. Radiat. Oncol. Biol. Phys. 2019, 108, S22–S23. [Google Scholar] [CrossRef]
  64. Goff, K.M.; Zheng, C.; Alonso-Basanta, M. Proton radiotherapy for glioma and glioblastoma. Chin. Clin. Oncol. 2022, 11, 46. [Google Scholar] [CrossRef] [PubMed]
  65. Kawabata, S.; Suzuki, M.; Hirose, K.; Tanaka, H.; Kato, T.; Goto, H.; Narita, Y.; Miyatake, S.I. Accelerator-based BNCT for patients with recurrent glioblastoma: A multicenter phase II study. Neuro-Oncol. Adv. 2021, 3, vdab067. [Google Scholar] [CrossRef] [PubMed]
  66. Shimizu, S.; Nakai, K.; Li, Y.; Mizumoto, M.; Kumada, H.; Ishikawa, E.; Yamamoto, T.; Matsumura, A.; Sakurai, H. Boron neutron capture therapy for recurrent glioblastoma multiforme: Imaging evaluation of a case with long-term local control and survival. Cureus 2023, 15, e33898. [Google Scholar] [CrossRef]
  67. Ocanto, A.; Torres, L.; Montijano, M.; Rincón, D.; Fernández, C.; Sevilla, B.; Gonsalves, D.; Teja, M.; Guijarro, M.; Glaría, L.; et al. MR-LINAC, a new partner in radiation oncology: Current landscape. Cancers 2024, 16, 270. [Google Scholar] [CrossRef]
  68. Detsky, J.; Chan, A.W.; Palhares, D.M.; Hudson, J.M.; Stewart, J.; Chen, H.; Das, S.; Lipsman, N.; Lim-Fat, M.J.; Perry, J.; et al. MR-Linac on-line weekly adaptive radiotherapy for high grade glioma (HGG): Results from the UNITED single arm phase II trial. Int. J. Radiat. Oncol. Biol. Phys. 2024, 120, S4. [Google Scholar] [CrossRef]
  69. Ballo, M.T.; Conlon, P.; Lavy-Shahaf, G.; Kinzel, A.; Vymazal, J.; Rulseh, A.M. Association of tumor treating fields (TTFields) therapy with survival in newly diagnosed glioblastoma: A systematic review and meta-analysis. J. Neurooncol. 2023, 164, 1–9. [Google Scholar] [CrossRef]
  70. Khagi, S.; Kotecha, R.; Gatson, N.T.N.; Jeyapalan, S.; Abdullah, H.I.; Avgeropoulos, N.G.; Batzianouli, E.T.; Giladi, M.; Lustgarten, L.; Goldlust, S.A. Recent advances in tumor treating fields (TTFields) therapy for glioblastoma. Oncologist 2025, 30, oyae227. [Google Scholar] [CrossRef]
  71. Rominiyi, O.; Vanderlinden, A.; Clenton, S.J.; Bridgewater, C.; Al-Tamimi, Y.; Collis, S.J. Tumour treating fields therapy for glioblastoma: Current advances and future directions. Br. J. Cancer 2021, 124, 697–709. [Google Scholar] [CrossRef]
  72. Szasz, A.M.; Arrojo Alvarez, E.E.; Fiorentini, G.; Herold, M.; Herold, Z.; Sarti, D.; Dank, M. Meta-analysis of modulated electro-hyperthermia and tumor treating fields in the treatment of glioblastomas. Cancers 2023, 15, 880. [Google Scholar] [CrossRef] [PubMed]
  73. Roussakow, S.V. Clinical and economic evaluation of modulated electrohyperthermia concurrent to dose-dense temozolomide 21/28 days regimen in the treatment of recurrent glioblastoma: A retrospective analysis of a two-centre German cohort trial with systematic comparison. BMJ Open 2017, 7, e017387. [Google Scholar] [CrossRef]
  74. Lee, S.Y.; Lorant, G.; Grand, L.; Szasz, A.M. The clinical validation of modulated electro-hyperthermia (mEHT). Cancers 2023, 15, 4569. [Google Scholar] [CrossRef]
  75. Ius, T.; Somma, T.; Pasqualetti, F.; Berardinelli, J.; Vitulli, F.; Caccese, M.; Cella, E.; Cenciarelli, C.; Pozzoli, G.; Sconocchia, G.; et al. Local therapy in glioma: An evolving paradigm from history to horizons (review). Oncol. Lett. 2024, 28, 440. [Google Scholar] [CrossRef]
  76. Mallick, S.; Gupta, S.; Amariyil, A.; Kunhiparambath, H.; Laviraj, M.A.; Sharma, S.; Sagiraju, H.K.R.; Julka, P.K.; Sharma, D.; Rath, G.K. Hypo-fractionated accelerated radiotherapy with concurrent and maintenance temozolomide in newly diagnosed glioblastoma: Updated results from phase II HART-GBM trial. J. Neurooncol. 2023, 164, 141–146. [Google Scholar] [CrossRef] [PubMed]
  77. Chidley, P.; Shanker, M.; Phillips, C.; Haghighi, N.; Pinkham, M.B.; Whittle, J.R.; Sia, J. Moderately hypofractionated versus conventionally fractionated radiation therapy with temozolomide for young and fit patients with glioblastoma: An institutional experience and meta-analysis of literature. J. Neurooncol. 2022, 160, 361–374. [Google Scholar] [CrossRef]
  78. Guo, L.; Li, X.; Chen, Y.; Liu, R.; Ren, C.; Du, S. The efficacy of hypofractionated radiotherapy (HFRT) with concurrent and adjuvant temozolomide in newly diagnosed glioblastoma: A meta-analysis. Cancer Radiother. 2021, 25, 182–190. [Google Scholar] [CrossRef] [PubMed]
  79. Klement, R.J.; Popp, I.; Kaul, D.; Ehret, F.; Grosu, A.L.; Polat, B.; Sweeney, R.A.; Lewitzki, V. Accelerated hyper-versus normofractionated radiochemotherapy with temozolomide in patients with glioblastoma: A multicenter retrospective analysis. J. Neurooncol. 2022, 156, 407–417. [Google Scholar] [CrossRef]
  80. Liao, G.; Zhao, Z.; Yang, H.; Li, X. Efficacy and safety of hypofractionated radiotherapy for the treatment of newly diagnosed glioblastoma multiforme: A systematic review and meta-analysis. Front. Oncol. 2019, 9, 1017. [Google Scholar] [CrossRef]
  81. Singh, R.; Lehrer, E.J.; Wang, M.; Perlow, H.K.; Zaorsky, N.G.; Trifiletti, D.M.; Bovi, J.; Navarria, P.; Scoccianti, S.; Gondi, V.; et al. Dose escalated radiation therapy for glioblastoma multiforme: An international systematic review and meta-analysis of 22 prospective trials. Int. J. Radiat. Oncol. Biol. Phys. 2021, 111, 371–384. [Google Scholar] [CrossRef]
  82. Andratschke, N.; Heusel, A.; Albert, N.L.; Alongi, F.; Baumert, B.G.; Belka, C.; Castellano, A.; Dhermain, F.; Erridge, S.; Grosu, A.; et al. ESTRO/EANO recommendation on reirradiation of glioblastoma. Radiother. Oncol. 2025, 204, 110696. [Google Scholar] [CrossRef]
  83. Lo Greco, M.C.; Milazzotto, R.; Liardo, R.L.E.; Acquaviva, G.; La Rocca, M.; Altieri, R.; Certo, F.; Barbagallo, G.M.; Basile, A.; Foti, P.V.; et al. Relapsing high-grade glioma from peritumoral zone: Critical review of radiotherapy treatment options. Brain Sci. 2022, 12, 416. [Google Scholar] [CrossRef] [PubMed]
  84. Preusser, M.; Kazda, T.; Le Rhun, E.; Sahm, F.; Smits, M.; Gempt, J.; Koekkoek, J.A.; Monti, A.F.; Csanadi, M.; Pitter, J.G.; et al. Lomustine with or without reirradiation for first progression of glioblastoma, LEGATO, EORTC-2227-BTG: Study protocol for a randomized phase III study. Trials 2024, 25, 366. [Google Scholar] [CrossRef]
  85. Chen, A.T.C.; Serante, A.R.; Ayres, A.S.; Tonaki, J.O.; Moreno, R.A.; Shih, H.; Gattás, G.S.; Lopez, R.V.M.; Dos Santos de Jesus, G.R.; de Carvalho, I.T.; et al. Prospective randomized phase 2 trial of hypofractionated stereotactic radiation therapy of 25 Gy in 5 fractions compared with 35 Gy in 5 fractions in the reirradiation of recurrent glioblastoma. Int. J. Radiat. Oncol. Biol. Phys. 2024, 119, 1122–1132. [Google Scholar] [CrossRef] [PubMed]
  86. Nabian, N.; Ghalehtaki, R.; Zeinalizadeh, M.; Balaña, C.; Jablonska, P.A. State of the neoadjuvant therapy for glioblastoma multiforme—Where do we stand? Neuro-Oncol. Adv. 2024, 6, vdae028. [Google Scholar] [CrossRef] [PubMed]
  87. Miyake, K.; Ogawa, T.; Fujii, T. STMO-15 Our therapeutic strategies for glioblastoma: Intraoperative support systems [intraoperative MRI, PET, 5-aminolevulinic acid (5-ALA)] and neoadjuvant chemotherapy. Neuro-Oncol. Adv. 2019, 1 (Suppl. 2), ii29. [Google Scholar] [CrossRef]
  88. Waqar, M.; Roncaroli, F.; Djoukhadar, I.; Akkari, L.; O’Leary, C.; Hewitt, L.; Forte, G.; Jackson, R.; Hessen, E.; Withington, L.; et al. Study protocol: Preoperative brain irradiation in glioblastoma (POBIG)—A phase I trial. Clin. Transl. Radiat. Oncol. 2023, 39, 100585. [Google Scholar] [CrossRef]
  89. Bihan, C.; Foscolo, S.; Boone, M.; Blonski, M.; Coutte, A.; Darlix, A.; Beauchesne, P.; Lefranc, M.; Lorgis, V.; Taillandier, L.; et al. Upfront bevacizumab and temozolomide or fotemustine before radiotherapy for patients with glioblastoma and severe neurological impairment at diagnosis. Case Rep. Oncol. 2012, 5, 530–536. [Google Scholar] [CrossRef]
  90. Balana, C.; De Las Penas, R.; Sepúlveda, J.M.; Gil-Gil, M.J.; Luque, R.; Gallego, O.; Carrato, C.; Sanz, C.; Reynes, G.; Herrero, A.; et al. Bevacizumab and temozolomide versus temozolomide alone as neoadjuvant treatment in unresected glioblastoma: The GENOM 009 randomized phase II trial. J. Neurooncol. 2016, 127, 569–579. [Google Scholar] [CrossRef]
  91. Jiang, H.; Zeng, W.; Ren, X.; Cui, Y.; Li, M.; Yang, K.; Elbaroody, M.; Lin, S. Super-early initiation of temozolomide prolongs the survival of glioblastoma patients without gross-total resection: A retrospective cohort study. J. Neurooncol. 2019, 144, 127–135. [Google Scholar] [CrossRef]
  92. Sun, Y.; Wang, Y.; Mu, S.; Wu, X.; Yu, S.; Wang, Z. Combining immunotherapy with radiotherapy for glioblastoma: Recent advances and challenges. Front. Oncol. 2025, 15, 1523675. [Google Scholar] [CrossRef]
  93. Long, G.V.; Shklovskaya, E.; Satgunaseelan, L.; Mao, Y.; da Silva, I.P.; Perry, K.A.; Diefenbach, R.J.; Gide, T.N.; Shivalingam, B.; Buckland, M.E.; et al. Neoadjuvant triplet immune checkpoint blockade in newly diagnosed glioblastoma. Nat. Med. 2025, 31, 1557–1566. [Google Scholar] [CrossRef] [PubMed]
  94. Baskaran, A.B.; Kozel, O.A.; Venkatesh, O.; Wainwright, D.A.; Sonabend, A.M.; Heimberger, A.B.; Lukas, R.V. Immune checkpoint inhibitors in glioblastoma IDHwt treatment: A systematic review. Cancers 2024, 16, 4148. [Google Scholar] [CrossRef] [PubMed]
  95. Armstrong, T.S.; Wefel, J.S.; Wang, M.; Gilbert, M.R.; Won, M.; Bottomley, A.; Mendoza, T.R.; Coens, C.; Werner-Wasik, M.; Brachman, D.G.; et al. Net clinical benefit analysis of Radiation Therapy Oncology Group 0525: A phase III trial comparing conventional adjuvant temozolomide with dose-intensive temozolomide in patients with newly diagnosed glioblastoma. J. Clin. Oncol. 2013, 31, 4076–4084. [Google Scholar] [CrossRef] [PubMed]
  96. Herrlinger, U.; Tzaridis, T.; Mack, F.; Steinbach, J.P.; Schlegel, U.; Sabel, M.; Hau, P.; Kortmann, R.D.; Krex, D.; Grauer, O.; et al. Lomustine-temozolomide combination therapy versus standard temozolomide therapy in patients with newly diagnosed glioblastoma with methylated MGMT promoter (CeTeG/NOA-09): A randomised, open-label, phase 3 trial. Lancet 2019, 393, 678–688. [Google Scholar] [CrossRef]
  97. Reardon, D.A.; Desjardins, A.; Peters, K.; Gururangan, S.; Sampson, J.; Rich, J.N.; McLendon, R.; Herndon, J.E.; 2nd Marcello, J.; Threatt, S.; et al. Phase II study of metronomic chemotherapy with bevacizumab for recurrent glioblastoma after progression on bevacizumab therapy. J. Neurooncol. 2011, 103, 371–379. [Google Scholar] [CrossRef]
  98. Wick, W.; Gorlia, T.; Bendszus, M.; Taphoorn, M.; Sahm, F.; Harting, I.; Brandes, A.A.; Taal, W.; Domont, J.; Idbaih, A.; et al. Lomustine and bevacizumab in progressive glioblastoma. N. Engl. J. Med. 2017, 377, 1954–1963. [Google Scholar] [CrossRef]
  99. Cruz Da Silva, E.; Mercier, M.-C.; Etienne-Selloum, N.; Dontenwill, M.; Choulier, L. A systematic review of glioblastoma-targeted therapies in phases II, III, IV clinical trials. Cancers 2021, 13, 1795. [Google Scholar] [CrossRef]
  100. Chinot, O.L.; Wick, W.; Mason, W.; Henriksson, R.; Saran, F.; Nishikawa, R.; Carpentier, A.F.; Hoang-Xuan, K.; Kavan, P.; Cernea, D.; et al. Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N. Engl. J. Med. 2014, 370, 709–722. [Google Scholar] [CrossRef]
  101. Friedman, H.S.; Prados, M.D.; Wen, P.Y.; Mikkelsen, T.; Schiff, D.; Abrey, L.E.; Yung, W.K.; Paleologos, N.; Nicholas, M.K.; Jensen, R.; et al. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J. Clin. Oncol. 2009, 27, 4733–4740. [Google Scholar] [CrossRef]
  102. Stupp, R.; Hegi, M.E.; Gorlia, T.; Erridge, S.C.; Perry, J.; Hong, Y.K.; Aldape, K.D.; Lhermitte, B.; Pietsch, T.; Grujicic, D.; et al. Cilengitide combined with standard treatment for patients with newly diagnosed glioblastoma with methylated MGMT promoter (CENTRIC EORTC 26071-22072 study): A multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2014, 15, 1100–1108. [Google Scholar] [CrossRef]
  103. Sanai, N.; Mehta, S.; Tien, A.-C.; Tovmasyan, A.; Harmon, J.; Kennedy, W.R.; Aziez, A.; Chalmers, A.J.; Khasraw, M.; Lim-Fat, M.J.; et al. A global phase 3, open-label, randomized 2-arm study comparing the clinical efficacy and safety of niraparib with temozolomide in adult participants with newly-diagnosed, MGMT unmethylated glioblastoma. J. Clin Oncol. 2025, 43 (Suppl. 16), TPS2096. [Google Scholar] [CrossRef]
  104. McFaline-Figueroa, J.R.; Sun, L.; Youssef, G.C.; Huang, R.; Li, G.; Kim, J.; Lee, E.Q.; Nayak, L.; Chukwueke, U.; Beroukhim, R.; et al. Neoadjuvant anti-PD1 immunotherapy for surgically accessible recurrent glioblastoma: Clinical and molecular outcomes of a stage 2 single-arm expansion cohort. Nat. Commun. 2024, 15, 10757. [Google Scholar] [CrossRef] [PubMed]
  105. Nassiri, F.; Patil, V.; Yefet, L.S.; Singh, O.; Liu, J.; Dang, R.M.A.; Yamaguchi, T.N.; Daras, M.; Cloughesy, T.F.; Colman, H.; et al. Oncolytic DNX-2401 virotherapy plus pembrolizumab in recurrent glioblastoma: A phase 1/2 trial. Nat. Med. 2023, 29, 1370–1378. [Google Scholar] [CrossRef]
  106. Weller, M.; Butowski, N.; Tran, D.D.; Recht, L.D.; Lim, M.; Hirte, H.; Ashby, L.; Mechtler, L.; Goldlust, S.A.; Iwamoto, F.; et al. Rindopepimut with temozolomide for patients with newly diagnosed, EGFRvIII-expressing glioblastoma (ACT IV): A randomised, double-blind, international phase 3 trial. Lancet Oncol. 2017, 18, 1373–1385. [Google Scholar] [CrossRef]
  107. Martucci, M.; Russo, R.; Giordano, C.; Schiarelli, C.; D’Apolito, G.; Tuzza, L.; Lisi, F.; Ferrara, G.; Schimperna, F.; Vassalli, S.; et al. Advanced magnetic resonance imaging in the evaluation of treated glioblastoma: A pictorial essay. Cancers 2023, 15, 3790. [Google Scholar] [CrossRef] [PubMed]
  108. Pineda, C.; Oleaga Zufiria, L.; Valduvieco Ruiz, I.; Pineda Losada, E.; Pujol Farré, T.; González Ortiz, S. RANO-2.0: Update to the response assessment for gliomas. Radiologia 2024, 41, 101621. [Google Scholar] [CrossRef]
  109. Banisharif, S.; Shahbazi-Gahrouei, D.; Akhavan, A.; Rasouli, N.; Shahbazi-Gahrouei, S. Determining the Optimum Tumor Control Probability Model in Radiotherapy of Glioblastoma Multiforme Using Magnetic Resonance Imaging Data Pre- and Post-Radiation Therapy. J. Res. Med. Sci. 2022, 27, 10. [Google Scholar] [CrossRef] [PubMed]
  110. Albert, N.L.; Galldiks, N.; Ellingson, B.M.; van den Bent, M.J.; Chang, S.M.; Cicone, F.; de Groot, J.; Koh, E.S.; Law, I.; Le Rhun, E.; et al. PET-based response assessment criteria for diffuse gliomas (PET RANO 1.0): A report of the RANO group. Lancet Oncol. 2024, 25, e29–e41. [Google Scholar] [CrossRef]
  111. Galldiks, N.; Lohmann, P.; Friedrich, M.; Werner, J.M.; Stetter, I.; Wollring, M.M.; Ceccon, G.; Stegmayr, C.; Krause, S.; Fink, G.R.; et al. PET imaging of gliomas: Status quo and quo vadis? Neuro Oncol. 2024, 26 (Suppl. 9), S185–S198. [Google Scholar] [CrossRef]
  112. Russo, G.; Stefano, A.; Alongi, P.; Comelli, A.; Catalfamo, B.; Mantarro, C.; Longo, C.; Altieri, R.; Certo, F.; Cosentino, S.; et al. Feasibility on the use of radiomics features of 11[C]-MET PET/CT in central nervous system tumours: Preliminary results on potential grading discrimination using a machine learning model. Curr. Oncol. 2021, 28, 5318–5331. [Google Scholar] [CrossRef] [PubMed]
  113. Verma, P.; Singh, B.K.; Sudhan, M.D.; Singh, R.K.; Bagul, S.D.; Chandak, A.R.; Soni, B.K.; Shelly, D.; Basu, S. 68 Ga-PSMA-11 PET/CT imaging in brain gliomas and its correlation with clinicopathological prognostic parameters. Clin. Nucl. Med. 2023, 48, e559–e563. [Google Scholar] [CrossRef] [PubMed]
  114. Zhu, M.; Li, S.; Kuang, Y.; Hill, D.L.; Tscheikner, M.E.; Alnawaf, H.; Koh, E.-S.; Malycha, P.; McLean, M.; Penniment, M.; et al. Artificial Intelligence in the Radiomic Analysis of Glioblastomas: A Review, Taxonomy, and Perspective. Front. Oncol. 2022, 12, 924245. [Google Scholar] [CrossRef] [PubMed]
Table 1. Survival patterns in glioblastoma multiforme by MGMT methylation status and therapy.
Table 1. Survival patterns in glioblastoma multiforme by MGMT methylation status and therapy.
InterventionMedian Survival–UnmethylatedMedian Survival–MethylatedConclusion
Surgery + RT11.8 months15.3 monthsRT alone improves OS in methylated tumors, but the benefit is limited.
Surgery + RT + TMZ12.7 months21.7 monthsThe addition of TMZ significantly increases OS in methylated tumors (p = 0.007).
Surgery + RT + TMZ + TTFields16.9 months31.6 monthsThe combination of TTFields with RT and TMZ offers the greatest OS benefit, especially in methylated tumors.
Legend: RT: Radiotherapy; TMZ: Temozolomide; TTFields: Tumor Treating Fields; OS: Overall Survival. Adapted from Roubil JG et al. [17], with data from Stupp et al. [12,16].
Table 2. Summary of studies on hypofractionation in patients with GBM.
Table 2. Summary of studies on hypofractionation in patients with GBM.
StudyPatientsTreatmentResultsConclusion
Roa et al.
(2004) [19]
N = 100
≥60 y.o.
KPS ≥ 50.
RT (60 Gy/6 weeks) vs. RT (40 Gy/3 weeks).
-
OS comparable (5.1 vs. 5.6 months)
-
Better tolerance
-
Lower use of post-treatment steroids in short RT
Short RT is effective and more comfortable
Perry et al.
(2017) [18]
N = 562
≥65 y.o.
RT (40 Gy/15 fr) ± concomitant and adjuvant TMZ.
-
Better OS with combination (9.3 mo vs. 7.6 m), especially in MGMT-methylated
-
More hematological adverse effects with TMZ
Short RT + TMZ improves OS.
Standard in eligible patients
Malmström et al. Nordic Trial
(2012) [22]
N = 342
≥60 y.o.
WHO 0–2
TMZ
vs.
RT 60 Gy/6 weeks
vs.
RT 34 Gy/2 weeks.
-
OS: TMZ 8.3 m/RT 6.0 mo/hypofractionated RT 7.5 mo
-
greater benefit in >70 years
-
MGMT-methylated: predictive marker of response to TMZ (OS 9.7 mo vs. 8.2 in RT)
-
TMZ and hypofractionated RT > standard RT
-
TMZ vs. hypofractionated RT in patients >70 years old, depending on MGMT-methylated. Individualized management
Roa et al.
IAEA Trial
(2015) [20]
N = 98
≥65 y.o.
and/or fragile
RT (25 Gy/5 fr) vs. RT (40.05 Gy/15 fr).Similar OS (7.9 vs. 6.4 mo).Hypofractionated RT is feasible and effective in frail patients
Minniti et al.
(2009) [21]
N = 43
≥70 y.o.
KPS ≥ 60
RT 30Gy/6 fr vs. adj. TMZ
-
OS 9.3 mo
-
PFS 6.3 mo
-
Best in KPS > 70
-
Acceptable toxicity
Combination is effective and safe in selected patients with limited prognosis
Legend: RT: Radiotherapy; TMZ: Temozolomide; KPS: Karnofsky Performance Status; WHO: World Health Organization scale; OS: Overall Survival; PFS: Progression-Free Survival; mo: Months; fr: Fractions.
Table 3. Summary of radiotherapy volumes for GBM.
Table 3. Summary of radiotherapy volumes for GBM.
Guideline/StudyGTVCTVPTVEdema Inclusion
EORTC (Stupp) [12]Tumor + cavityCTV1: GTV + edema + 20 mm
CTV2: GTV + 25 mm
PTV = CTV + 3–5 mmYes, included in initial phase
RTOG/NRG
(2019) [14]
Phase 1: tumor + edema; Phase 2: tumor + cavityCTV1: GTV1 + 20 mm
CTV2: GTV2 + 20 mm
PTV = CTV + 3–5 mmYes, included in initial phase
ESTRO-EANO 2023 [25]Surgical cavity + post-surgical T1 enhancementGTV + 15 mm (adjusted to anatomy)CTV + individual margin (usually ≤3 mm with IGRT)Not systematically included
ESTRO-ACROP 2016 [26]Cavity + residual tumorGTV + 15–20 mm, adjusted to anatomical barriersCTV + 3–5 mmNot systematically included
Minniti et al. (2010) [28]Surgical cavity + post-surgical T1 enhancementCTV1: GTV + 2 cm
CTV2: GTV + 1 cm
CTV + 3mmNot included
Chang et al.
(2007) [29]
Cavity + T1 tumorCTV1: GTV + 20 mm
CTV2: GTV + 5 mm
CTV + 5 mmNot included
ASTRO 2025
(1-phase) [11]
Surgical cavity + post-surgical T1 enhancementGTV + 10–20 mm (adjusted to anatomy, including edema is optional)PTV = CTV + 3–5 mmOptional.
ASTRO 2025
(2-phase) [11]
Phase 1: cavity + T1 + T2/FLAIR enhancement
Phase 2: cavity + T1 enhancement
CTV1: GTV1 + 10–20 mm
CTV2: GTV2 + 10–20 mm (adjusted to anatomy)
PTV = CTV + 3–5 mmYes, included in initial phase; not in phase 2
MDACC
Kumar et al.
(2020)
[27]
Cavity + T1 enhancementInitial GTV + 2 cm, boost GTV + 5 mmCTV + 5 mmNot included
Legend: GTV: Gross Tumor Volume; CTV: Clinical Target Volume; PTV: Planning Target Volume; IGRT: Image-Guided Radiotherapy; MDACC: MD Anderson Cancer Center.
Table 4. Limiting factors of radiotherapy in the treatment of GBM.
Table 4. Limiting factors of radiotherapy in the treatment of GBM.
CategoryLimitationDescription/Evidence
TechnicalLocal recurrence80–90% of recurrences occur within 2 cm of the irradiated field due to diffuse infiltration, even with IMRT, VMAT, or proton therapy [32].
Dosimetric constraintsLimits such as <54 Gy to the brainstem and optic chiasm restrict dose escalation; proton therapy minimizes irradiated volumes [25].
Acute and late toxicityFatigue, radiation necrosis, and cognitive deficits; brain volumes (V20Gy, V40Gy) increase neurotoxicity, reducible with IMRT, VMAT, and proton therapy [25].
Delay in RT initiationDelays >6 weeks worsen OS and PFS; moderate delays (~6 weeks) may benefit patients with residual disease [33,34].
Cost and adherence of new technologiesTumor Treating Fields (TTFields) extend PFS by 2.7 months (6.7 vs. 4.0 months), limited by cost and adherence (≥18 h/day) [35].
BiologicalTumor infiltrationThe diffuse nature of GBM allows tumor cells to escape the radiation field [32].
Tumor hypoxiaTumor hypoxia, by activating HIF-1α, reduces RT efficacy by promoting cell survival [36]. Mitigated with modulated electrohyperthermia (mEHT) or hypoxia-sensitizing agents; high-linear energy transfer (LET, energy deposited per unit length) radiation (e.g., carbon ions; alpha particles) is promising but investigational [37,38].
Cellular radioresistanceTumor stem cells and pathways like MGMT (O6-methylguanine-DNA methyltransferase), EGFR amplification (57% per TCGA [The Cancer Genome Atlas]), PTEN mutations (40% per TCGA), and CDKN2A/B deletions (60% per TCGA) drive resistance; PARP inhibitors (e.g., veliparib) and other radiosensitizers show promise by inhibiting DNA repair [36,39,40]. See Table 5 for additional molecular determinants.
EGFR amplificationIn 57% per TCGA, activates PI3K/Akt and RAS/RAF/MAPK, conferring resistance; PTEN mutations (40% per TCGA) enhance this pathway; inhibitors like erlotinib have limited benefits [36,40,41].
SVZ as a reservoirThe SVZ (subventricular zone), with mutated stem cells (TERT promoter mutation >70% per TCGA, PTEN, TP53, EGFR), drives regrowth; irradiating the SVZ with doses ≥56 Gy (ipsilateral) or ≥50 Gy (contralateral) does not improve PFS or OS [40,42,43].
RT-induced lymphopeniaExtensive irradiation causes grade 3+ lymphopenia (14% with protons vs. 39% with photons), limiting immunotherapy efficacy [44].
Microenvironment and cancer neuroscienceImmunosuppressive microenvironment (e.g., macrophages/microglia supporting tumor growth) and neuronal crosstalk (e.g., synapses promoting proliferation/invasion) enhance resistance [45].
ClinicalPseudoprogressionAffects 30-40% of patients with methylated MGMT after TMZ, complicating radiological assessment up to 12 weeks [46,47].
Lack of clinical impact of biomarkers and advanced imagingMultimodal imaging (multiparametric MRI; amino acid PET) enables RT personalization, achieving an mOS of 23 months in a phase I trial [48]. Biomarkers (EGFR, PTEN, and TERT) allow patient stratification but, except for MGMT-methylated, do not improve OS in phase III trials [49].
Lack of consistent benefits from combined therapiesTargeted therapies and immunotherapies do not improve OS, though they extend PFS in nGBM [50].
Legend: PFS: Progression-Free Survival; mOS: Median Overall Survival; IMRT: Intensity-Modulated Radiotherapy; VMAT: Volumetric Modulated Arc Therapy; PT: Proton Therapy; MGMT: O6-Methylguanine-DNA Methyltransferase; EGFR: Epidermal Growth Factor Receptor; PTEN: Phosphatase and Tensin Homolog; TERT: Telomerase Reverse Transcriptase; TP53: Tumor Protein p53; SVZ: Subventricular Zone; HIF-1α: Hypoxia-Inducible Factor 1-Alpha; nGBM: Newly Diagnosed Glioblastoma Multiforme; RT: Radiotherapy; TMZ: Temozolomide; MRI: Magnetic Resonance Imaging; PET: Positron Emission Tomography. TCGA: The Cancer Genome Atlas.
Table 5. Molecular biomarkers in RT for GBM.
Table 5. Molecular biomarkers in RT for GBM.
BiomarkerFrequency (IDH-wt)Impact on RTTherapeutic Status
MGMT-methylated~45%Greater sensitivity to RT + TMZ
(beneficial prognostic marker)
Standard treatment with TMZ. ESCAT I [12,51,52]
IDH1/2 Mutation0% (defines IDH-wt)Greater radiosensitivity and better prognosis in IDH-mutant astrocytoma grade 4 (rare, ~10% of grade 4 gliomas)Favorable stratification. ESCAT I [53]
Amplified EGFR57% (per TCGA)Activates PI3K/AKT; resistance to RTInhibitors without relevant clinical efficacy. ESCAT IIIA [54]
PTEN mutation/loss~40% (per TCGA)PI3K/AKT pathway; promotes resistance to RTNo approved effective therapies [40,55]
PIK3CA mutation~10% (per TCGA)Stimulates cellular survival signalsNo approved effective therapies [40,55]
Amplified CDK4/6~15% (per TCGA)Stimulates cell cycle progressionInhibitors under clinical study [40,55]
CDKN2A/B deletion~50% (per TCGA)Loss of cell cycle control; poor prognosisNo effective targeted therapies [49,56]
TERT mutation>70% (per TCGA)Uncertain impact; possible role in immune evasionUnder investigation as an immunotherapeutic target [3,57]
Legend: IDH: Isocitrate Dehydrogenase; wt: Wild-Type; MGMT: O6-Methylguanine-DNA Methyltransferase; RT: Radiotherapy; TMZ: Temozolomide; ESCAT: ESMO Scale for Clinical Actionability of Molecular Targets; EGFR: Epidermal Growth Factor Receptor; PTEN: Phosphatase and Tensin Homolog; PI3K/AKT: Phosphatidylinositol 3-Kinase/Protein Kinase B; CDK4/6: Cyclin-Dependent Kinases 4 and 6; TERT: Telomerase Reverse Transcriptase; CDKN2A/B: Cyclin-Dependent Kinase Inhibitor 2A/B; TCGA: The Cancer Genome Atlas.
Table 6. Technological advances in therapies against GBM.
Table 6. Technological advances in therapies against GBM.
TechniqueDescriptionmOS/PFSEvidenceResults PendingLimitations
PTFocused dose delivery (Bragg peak, spread-out Bragg peak [SOBP], intensity-modulated proton therapy [IMPT], RBE 1.1)nGBM: mOS 21–24 months; PFS 6.6–8.9 monthsPhase II (NCT01854554, n = 67; PT vs. XRT); reduces grade ≥ 3 lymphopenia (14% vs. 39%, p = 0.024), fatigue (24% vs. 58%, p = 0.05), toxicity grade ≥ 2 (0.35 vs. 1.15, p = 0.02), V5–V40 [44,62,63,64]NRG-BN001 (NCT02179086): Phase II trial, photon arm (75 Gy/30 fractions) improves PFS, mOS 18.7 months; proton arm results pending,
in nGBM [63,64]
Cost, accessibility
CIRTHigh energy transfer, RBE 2.5–5nGBM: mOS 18 months; rGBM: mOS 8 monthsnGBM: CIRT boost (18 GyE/6 fx) or with TMZ (retrospective randomized phase II); rGBM: 45 GyE/15 fx (non-randomized comparative) [37]CINDERELLA (NCT01166308): Phase I/II, CIRT vs. FSRT in rGBM; CLEOPATRA (NCT01165671): Phase II randomized, CIRT vs. proton boost in nGBM [37]Cost, limited centers. *
BNCTSelective damage with boron-10 (L-BPA, BSH); planning
with 18F-BPA PET
nGBM: mOS 25.7 months (with surgery + TMZ); rGBM: mOS 18.9 monthsnGBM: Surgery, BNCT (~40 Gy-Eq) and TMZ, without conventional RT; rGBM: Non-randomized Phase II (JG002), minimum 39.8 Gy-Eq [65,66]Under investigation, non-randomized
[65,66]
Toxicity (cerebral edema, hyperamylasemia, alopecia); infrastructure. *
MRgRTDaily adaptation with T1/T2 MRInGBM: mOS 18.5 months, PFS 11.6 months (long course); marginal failure 4.1%Non-randomized Phase II UNITED (NCT04726397, n = 98; CTV 5 mm, 60 Gy/30 fx) [67,68]UNITED2 (NCT05565521): Phase II non-randomized, 40 Gy/15 fx + boost 52.5 Gy/15 fx, PFS at 6 months [67]Cost, evidence in development. *
TTFieldsAlternating electric fields (200 kHz, 1–3 V/cm); with TMZ + RTnGBM: mOS 20.9 months, PFS 6.7 months (EF-14)
rGBM: mOS 10.3 months (EF-11)
nGBM: mOS 24.8 months, PFS 12.0 months, 1-year survival 82.61% (2-THE-TOP)
Phase III EF-14 (NCT00916409, n = 695; TTFields+TMZ in nGBM, HR 0.63, p < 0.001); phase III EF-11 (NCT00379470, TTFields in rGBM); phase II 2-THE-TOP (NCT03405792, TTFields + TMZ + pembrolizumab in nGBM) [69,70,71]Phase III TRIDENT (NCT04471844): RT/TMZ in nGBM; EF-41 (NCT06556363): TMZ+pembrolizumab + TTFields in nGBM [69,70,71]Cost, adherence, dermatitis; NICE does not endorse it due to cost-effectiveness, EF-14 biases (unblinded, selected patients)
mEHTThermal radiosensitization (13.56 MHz, 40–43 °C); immunogenic potentialnGBM: 1-year survival 73.33%; rGBM: mOS 7.7 months, 1-year survival 37.33%Observational studies 2006–2018 (n = 450); 1 nGBM study, 6 rGBM studies (with ddTMZ); phase I for safety; no RCTs
[72,73,74]
In research, non-randomized [72,73,74]Weak evidence, not in guidelines, few centers. *
LITTMRI-guided laser thermal ablationrGBM: mOS ~8–12 monthsPhase I/II in rGBM; comparable to re-surgery in unifocal lobar rGBM; no RCTs [75]In research, non-randomized [75]Small focal lesions, no RCTs, post-procedural edema. *
MHTAMF-guided hyperthermia with magnetic nanoparticles (40–45°C)rGBM: mOS not reportedPhase I/II in rGBM with RT; Proven safety and feasibility; no RCTs [75]In research, non-randomized [75]No RCTs, technical challenges (MNP, thermometry), few centers. *
Legend: AMF: Alternating magnetic field; BSH: Sodium borocaptate; CIRT: Carbon ion radiotherapy; ddTMZ: Dense dose temozolomide; FSRT: Fractionated stereotactic radiotherapy; GyE: Gray equivalent; HR: Hazard ratio; IMPT: Intensity-modulated proton therapy; L-BPA: L-4-boronophenylalanine; LITT: Laser interstitial thermal therapy; mEHT: Modulated electrohyperthermia; MHT: Magnetic hyperthermia; MNP: Magnetic nanoparticles; MRgRT: Magnetic resonance-guided radiotherapy; RBE: Relative biological effectiveness; RCTs: Randomized clinical trials; SOBP: Spread-out Bragg peak; V5–V40: Volume of tissue receiving 5 to 40 Gy of radiation; XRT: Photon radiotherapy. * Note: non-randomized data limit comparisons with photon-based radiotherapy.
Table 7. Modified fractionation schedules.
Table 7. Modified fractionation schedules.
Schedule/
Description/
Indications
mOS/PFSEvidenceOngoing TrialsLimitations
Hypofractionation: 50–60 Gy/20 fractions with TMZ, primarily in younger patients (≤65 years, KPS ≥ 70)mOS: 26.5 months, PFS: 13.2 months [76]; mOS: 19.8 months, PFS: 7.7 months [77]; 12-month OS: 71.3%, 12-month PFS: 40.8% (various ages) [78]HART-GBM trial (phase II, 60 Gy/20 fx vs. 60 Gy/30 fx, n = 83, with TMZ, patients aged 16–65 years) [76]; Institutional study (50 Gy/20 fx vs. 60 Gy/30 fx, n = 41, with TMZ, patients < 65 years) [77]; Meta-analysis (n = 484, phase I/II and retrospective, various ages) [78]; Meta-analysis (n not specified, phase II/III, HFRT vs. CFRT, various ages) [80]SAGA (NCT05781321, randomized phase II, 5–10 fx photons guided by [18F]-FDOPA PET, evaluating survival, cost-effectiveness, and failure patterns, patients ≥ 18 years, ClinicalTrials.gov)Grade ≥ 3 radionecrosis (6.7% HFRT, 7.7% CFRT), tumor heterogeneity [77,80]
Hyperfractionation: 37 × 1.6 Gy or 30 × 1.8 Gy bid with TMZ, experimentalNo clear benefit (mOS 14.9 vs. 16.9 months, p = 0.26) [79]Retrospective analysis (HFRT vs. NFRT, n = 484, with TMZ) [79]; Review (variable dose, with TMZ) [36] Lack of efficacy; moderate toxicity [36,79]
Dose escalation: 75 Gy/30 fx (IMRT/PT) with TMZ or 16.8–24.8 GyE boost (CIRT), selected patients (KPS ≥ 70)Photon arm:
mOS: 18.7 months, improved PFS (60); mOS: 18 months (CIRT boost) [37]
NRG-BN001 phase II trial (75 Gy/30 fx vs. 60 Gy/30 fx, n = 299, with TMZ, photons, preliminary results improve PFS, mOS 18.7 months)
[63]; CIRT retrospective study (16.8–24.8 GyE/8 fx after 50 Gy photons, n = 32) [37]
NRG-BN001 (PT arm, randomized phase II) [63]; CLEOPATRA (CIRT boost, randomized phase II) [37]Grade ≥ 3 radionecrosis (up to 29%), tumor heterogeneity, no mOS improvement [63,81]
Legend: bid: Twice daily; CIRT: Carbon ion radiotherapy; GyE: Gray equivalent.
Table 8. Reirradiation.
Table 8. Reirradiation.
ModalityDescriptionIndicationsMOS/
PFS
EvidenceTrialsLimitations
HFRT/
CFRT
35 Gy/10 fx or 36 Gy/18 fx ± BEV (CTV: GTV + ≤5 mm, adjusted to anatomical barriers; PTV: CTV + ≤3 mm) [82]KPS > 60, recurrence > 6 months, volume < 35 cm3 (ESTRO/EANO 2025) [82]; KPS ≥ 70, volume ≤ 6 cm3 (ASTRO 2025), multidisciplinary discussion [11]mOS 7–12 months, PFS 3–6
months
[82]
NRG Oncology/RTOG 1205 (phase II, HFRT + BEV, mOS 10.1 months) [82]LEGATO (phase III, lomustine ± HFRT)
[84]
Radionecrosis (~5%), lack of phase III trials [82]
SRS/
HSRT
SRS: 12–15 Gy/1 fx (CTV: GTV, usually without margin; PTV: CTV + 0–1 mm); HSRT: 25 Gy/5 fx (CTV: GTV, without margin; PTV: CTV + 3 mm) [82,83,85]SRS: volume < 10 cm3; HSRT: volume ≤ 150 cm3 (median 55 cm3) [82,83,85]mOS 9–11 months, PFS 5–6 months [82,83,85]Retrospective trials (SRS, volume < 12.5 cm3) [82]; Phase II, HSRT 25 Gy/5 fx, mOS 9.2 months, PFS 4.9 months; 35 Gy/5 fx shows no improvement in PFS (4.9 vs. 5.2 months) or OS (9.2 vs. 10 months) [85] Radionecrosis (<3.5% if volume <12.5 cm3 [82]; ~25% in HSRT [85]), phase II escalation from 25 Gy/5 fx to 35 Gy/5 fx does not improve PFS (4.9 vs. 5.2 months) or OS (9.2 vs. 10 months) [85]
CIRT/PTCIRT: 45 Gy RBE/15 fx; PT: 33–46.2 Gy variable (CTV: GTV + ≤3 mm, adjusted to anatomical barriers; PTV: CTV + ≤3 mm) [37,64,83]Selected patients [37,64,83]mOS 7.8–19.4 months, PFS 5.5–13.9 months [37,64,83]Retrospective studies (CIRT, PT) [83]; CIRT 45 Gy RBE/15 fx, mOS 8.0 months vs. photons [37]; PT 33–46.2 Gy, mOS 7.8–19.4 months, low toxicity [64]CINDERELLA (phase I/II, CIRT vs. FSRT) [64]Cost, accessibility, limited prospective data, toxicity not reported
Legend: BEV: Bevacizumab; CIRT: Carbon ion radiotherapy; fx: Fraction; HSRT: Hypofractionated stereotactic radiotherapy; PT: Proton therapy; RBE: Relative biological effectiveness; SRS: Stereotactic radiosurgery.
Table 9. Neoadjuvant therapy studies in GBM.
Table 9. Neoadjuvant therapy studies in GBM.
Preoperative Neoadjuvant Therapy for GBM
ModalityDescriptionmOS/PFSTrial/
Evidence
Limitations
Preoperative RTSRS (6–14 Gy/1 fx)Not reportedPOBIG (phase I) [88]No phase III trials
RT + Preoperative TMZRT + Preoperative TMZNot reportedPARADIGMA (phase II, NCT03480867) [86]Pending results
BEVBEV (10 mg/kg) preoperativemOS: 15.7 months, PFS: 10.1 monthsMiyake et al. (phase II, n = 12) [87]Small sample size; limited data on toxicity
PembrolizumabPembrolizumab (ICI, 200 mg every 3 weeks) pre-surgerymOS: 13.7 months(phase II, rGBM) [92]Small sample size, immunological toxicity, dose heterogeneity, potential influence of steroids and bevacizumab
Triple ImmunotherapyNivolumab + ipilimumab + relatlimab pre-surgeryNo recurrence at 17 months (n = 1)GIANT (phase I, nGBM, NCT06816927) [93]Single case; preliminary data
Postoperative Neoadjuvant Systemic Therapy Prior to Standard Chemoradiotherapy for Unresectable or Inoperable GBM
ModalityDescriptionmOS/PFSTrial
/Evidence
Limitations
TMZ + BEVTMZ (75 mg/m2) + BEV (10 mg/kg) pre-RTmOS: 12.5 months, PFS: 7.4–8.6 monthsBihan et al. (retrospective, n = 8) [89]
Balana et al. (phase II, n= 102) [90]
Intracranial hemorrhages *; increased toxicity
Postoperative Neoadjuvant Systemic Therapy Prior to Chemoradiotherapy for Resectable GBM
ModalityDescriptionmOS/PFSTrial/EvidenceLimitations
TMZTMZ (75 mg/m2 daily) <7 days post-surgery and extended adjuvant (150–200 mg/m2 until progression), before CRT (60 Gy/20 fx in MAGMA)mOS: 23 months, PFS: 11.5 monthsJiang et al. (retrospective, n = 375); MAGMA (phase III) [91] **Hematological toxicity, MAGMA pending
Legend: BEV: Bevacizumab; fx: Fraction; SRS: Stereotactic radiosurgery. * Note: Intracranial hemorrhages primarily associated with BEV (4.2% in Balana et al.) [90]. ** Note: Greater benefit in MGMT-methylated cases (HR 0.60, Jiang et al.) [91].
Table 10. Immunotherapy, targeted therapies, and chemotherapy.
Table 10. Immunotherapy, targeted therapies, and chemotherapy.
CategoryModalitymOS/PFSEvidenceNotes
Chemotherapy
(nGBM)
Standard TMZmOS 14.6 months, mPFS 6.9 monthsEORTC/NCIC CE.3 [12]Standard treatment
Intensive TMZNo improvement in mOS or PFSRTOG 0525 [95]Not recommended
Lomustine + TMZmOS 48.1 monthsCeTeG/NOA-09 [96]MGMT-methylated
Chemotherapy
(rGBM)
Metronomic TMZPFS-6 24% (1st recurrence); PFS-6 4.4% (post-BEV)Phase II [97]Limited efficacy, especially after bevacizumab
LomustinemPFS 1.5 monthsEORTC 26101 [98,99]Limited efficacy; alone or with bevacizumab
Targeted Therapies (nGBM)BVZmOS 16.8 months, mPFS 10.6 monthsAVAglio, RTOG 0825 [99,100,101]Not recommended as initial treatment; see Table 9 for neoadjuvant use
Cilengitide + TMZNo improvement SG/PFSCENTRIC, CORE [99,102]Not recommended
PARP inhibitors (e.g., veliparib)6-month PFS 46% (95% CI: 36–57%) vs. 31% (95% CI: 18–46%) in nGBM with unmethylated MGMT; no mOS benefit (12.7 vs. 12.8 months)Phase II VERTU, preclinical synergy with RT/TMZ 36Promising for 6-month PFS in nGBM with unmethylated MGMT, requires phase III confirmation, limited by tumor heterogeneity
PARP inhibitor (niraparib) vs. TMZEmerging (phase 3 for MGMT unmethylated)Phase 3 Gliofocus [103]
(NCT06388733)
Ongoing; potential alternative to TMZ in unmethylated cases; PFS/OS endpoints
Targeted Therapies (rGBM)BVZmOS 9.2 months, mPFS 4.2 monthsBRAIN, EORTC 26101, BELOB [98,99,101]Recommended in symptomatic relapse; see Table 9 for neoadjuvant use
RegorafenibmOS 7.4 months, mPFS 2.0 monthsREGOMA [99]Limited efficacy in rGBM
BRAF/MEK inhibitorsPartial answersBasket trials [99]Compassionate use or use in clinical trials
IDH inhibitors (vorasidenib)No data in GBMPhase I [99]In research for IDH-mutant gliomas
Erlotinib, EverolimusIneffectivePhase II [41,99]Not recommended
Antibody-drug conjugates (ADCs)Emerging (e.g., phase I/II for EGFR-targeted)Preclinical/early trials [45]Promising for targeted delivery, but no phase III data; potential synergy with RT
IT
(rGBM)
ICI: NivolumabmOS 9.8 monthsCheckMate 143 [3,92]No OS gain over BEV
ICI: PembrolizumabmOS 13.8 months, mPFS 3.3 months, PFS-6 19.5%Phase II (NCT02852655) [92,104]Phase II, neoadjuvant to surgery; benefit in subgroups; see Table 9
ACT: CAR-T, TILs, LAKmOS 20.5 months, 1 RCPhase I [92]Phase I, preliminary data
Vaccines: DCVax-LmOS 13.2 monthsPhase III [92]Phase III, without RT; benefit in mOS, non-standard
OV: DNX-2401, G47Δ, PVSRIPOmOS 12.5-20.2 monthsPhase I/II [92,105]Preliminary results from CAPTIVE (DNX-2401 + pembrolizumab, ongoing)
Cytokines: L19TNF + lomustinemPFS 43.3 weeks[92]Phase I, preliminary data
IT
(nGBM)
ICI: Nivolumab + RTmOS 13.4 monthsCheckMate 498 [94]No OS gain over TMZ + RT; MGMT not methylated
ICI: Nivolumab + TMZ + RTmOS 28.9 monthsCheckMate 548 [94]No OS gain over TMZ + RT; MGMT-methylated
Vaccines: DCVax-LmOS 19.3 monthsPhase III [92]Phase III, with RT + TMZ; benefit in mOS, non-standard
Vaccines: Rindopepimut + TMZmOS 20.0 monthsACT IV [92,106]Phase III, no improvement in OS; not recommended for EGFRvIII+
Cytokines: IFN-α + TMZmOS 26.7 monthsPhase III [92]Phase III, adjuvant after RT; benefit in mOS, non-standard
Triple ITNo recurrence at 17 monthsGIANT, ongoing [93]Neoadjuvant to surgery; single case; ongoing trials
Legend: ACT: Adoptive cellular therapies; BEV: Bevacizumab; ICI: Immune checkpoint inhibitors; IT: Immunotherapy; OV: Oncolytic viruses; PFS-6: 6-month progression-free survival.
Table 11. Advanced imaging and theragnostic.
Table 11. Advanced imaging and theragnostic.
ModalityApplicationPerformanceLimitations
Multiparametric MRIDiagnosis, recurrence, pseudoprogressionHigh sensitivity, RANO 2.0; DSC (90% sensitivity, 88% specificity), DWI (ADC >1200 × 10−6 mm2/s), MRS (low Cho/Cr and Cho/NAA) for pseudoprogression [47,107,108]Cost, need for specialized interpretation
PET 18F-FETTheranostics (general)High specificity (~80–90%), PET RANO 1.0
[110,111]
Cost, accessibility
PET [11C]-METDiagnosis, recurrence/
pseudoprogression
~95% sensitivity/specificity for grading; high accuracy for recurrence [111,112]Short half-life, accessibility
PET [18F]F-DOPADiagnosis, recurrence/
pseudoprogression
92% sensitivity, 75% specificity for recurrence
[110]
Cost, need for additional studies
PET [18F]FACBCDiagnosis, recurrence/
pseudoprogression
90% sensitivity, 83% specificity for recurrence
[111]
Need for further studies, accessibility
PET [68Ga]-PSMA-11Diagnosis, recurrence/
pseudoprogression
High uptake in high-grade gliomas [113]Need for further studies
Multiparametric MRI/PET-guided RTPersonalized RT with dose escalationmOS 23 months in a phase I trial using multiparametric MRI and [18F]-FDOPA PET; no OS benefit in phase III trials [48]Cost, accessibility, need for phase III validation
Theranostics [131I]-IPATreatment, evaluationmOS 16 months in rGBM [111,113]Limited BBB penetration, need for validation
Theranostics (general)Treatment, evaluationOngoing trials (e.g., [177Lu]-PSMA, [177Lu]-6A10, [177Lu]-NeoB) [112,113]Limited BBB penetration, need for validation
Legend: ADC: Apparent diffusion coefficient; BBB: Blood–brain barrier; Cho: Choline; Cr: Creatine; DSC: Dynamic susceptibility contrast perfusion; DWI: Diffusion-weighted imaging; MRS: Magnetic resonance spectroscopy; NAA: N-acetyl aspartate; RANO 2.0: 2023 criteria for glioma evaluation; PET RANO 1.0: Criteria for amino acid PET.
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

Fernández, C.; Ciérvide, R.; Díaz, A.; Garrido, I.; Couñago, F. Radiotherapy in Glioblastoma Multiforme: Evolution, Limitations, and Molecularly Guided Future. Biomedicines 2025, 13, 2136. https://doi.org/10.3390/biomedicines13092136

AMA Style

Fernández C, Ciérvide R, Díaz A, Garrido I, Couñago F. Radiotherapy in Glioblastoma Multiforme: Evolution, Limitations, and Molecularly Guided Future. Biomedicines. 2025; 13(9):2136. https://doi.org/10.3390/biomedicines13092136

Chicago/Turabian Style

Fernández, Castalia, Raquel Ciérvide, Ana Díaz, Isabel Garrido, and Felipe Couñago. 2025. "Radiotherapy in Glioblastoma Multiforme: Evolution, Limitations, and Molecularly Guided Future" Biomedicines 13, no. 9: 2136. https://doi.org/10.3390/biomedicines13092136

APA Style

Fernández, C., Ciérvide, R., Díaz, A., Garrido, I., & Couñago, F. (2025). Radiotherapy in Glioblastoma Multiforme: Evolution, Limitations, and Molecularly Guided Future. Biomedicines, 13(9), 2136. https://doi.org/10.3390/biomedicines13092136

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