Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (453)

Search Parameters:
Keywords = Brain Tumours

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 8171 KiB  
Article
Improving the Treatment of Brain Gliomas Through Small-Particle-Size Paclitaxel-Loaded Micelles with a High Safety Profile
by Bohan Chen, Liming Gong, Jing Feng, MongHsiu Song, Mingji Jin, Liqing Chen, Zhonggao Gao and Wei Huang
Pharmaceutics 2025, 17(8), 965; https://doi.org/10.3390/pharmaceutics17080965 - 25 Jul 2025
Viewed by 288
Abstract
Background/Objectives: Paclitaxel (PTX) is widely used in the treatment of a variety of solid tumours due to its broad-spectrum anti-tumour activity, but its use in brain gliomas is limited by insufficient blood–brain tumour barrier (BBTB) penetration and systemic toxicity. The aim of [...] Read more.
Background/Objectives: Paclitaxel (PTX) is widely used in the treatment of a variety of solid tumours due to its broad-spectrum anti-tumour activity, but its use in brain gliomas is limited by insufficient blood–brain tumour barrier (BBTB) penetration and systemic toxicity. The aim of this study was to develop a Solutol HS-15-based micellar nanoparticle (PSM) to enhance the brain glioma targeting of PTX and reduce toxicity. Methods: PSMs were prepared by solvent injection and characterised for particle size, encapsulation rate, haemolysis rate and in vitro release properties. A C6 in situ glioma mouse model was used to assess the brain targeting and anti-tumour effects of the PSM by in vivo imaging, tissue homogenate fluorescence analysis and bioluminescence monitoring. Meanwhile, its safety was evaluated by weight monitoring, serum biochemical indexes and histopathological analysis. Results: The particle size of PSMs was 13.45 ± 0.70 nm, with an encapsulation rate of 96.39%, and it demonstrated excellent cellular uptake. In tumour-bearing mice, PSMs significantly enhanced brain tumour targeting with a brain drug concentration 5.94 times higher than that of free PTX. Compared with Taxol, PSMs significantly inhibited tumour growth (terminal luminescence intensity <1 × 106 p/s/cm2/Sr) and did not cause significant liver or kidney toxicity or body weight loss. Conclusions: PSMs achieve an efficient accumulation of brain gliomas through passive targeting and EPR effects while significantly reducing the systemic toxicity of PTX. Its simple preparation process and excellent therapeutic efficacy support its use as a potential clinically translational candidate for glioma treatment. Full article
Show Figures

Figure 1

18 pages, 1243 KiB  
Article
Incidence and Clinical Features of Pseudoprogression in Brain Metastases After Immune-Checkpoint Inhibitor Therapy: A Retrospective Study
by Chris W. Govaerts, Miranda C. A. Kramer, Ingeborg Bosma, Frank A. E. Kruyt, Frederike Bensch, J. Marc C. van Dijk, Mathilde Jalving and Anouk van der Hoorn
Cancers 2025, 17(15), 2425; https://doi.org/10.3390/cancers17152425 - 22 Jul 2025
Viewed by 236
Abstract
Background: Pseudoprogression is known to occur after immune-checkpoint inhibitor (ICI) therapy in brain metastasis and can complicate clinical decision-making. Still, its incidence, timing, and clinical presentation remain unclear. A retrospective cohort study in melanoma and non-small cell lung cancer brain metastasis patients was [...] Read more.
Background: Pseudoprogression is known to occur after immune-checkpoint inhibitor (ICI) therapy in brain metastasis and can complicate clinical decision-making. Still, its incidence, timing, and clinical presentation remain unclear. A retrospective cohort study in melanoma and non-small cell lung cancer brain metastasis patients was conducted to address this. Materials and Methods: Brain metastasis patients showing progression on MRI according to response assessment in neuro-oncology brain metastases criteria after starting ICI therapy were included, irrespective of prior irradiation. Lesions were classified as tumour progression (TP) or pseudoprogression based on three-month radiological follow-up or histopathology. TP was assigned if progression was again shown at three months. Pseudoprogression was assigned if lesions showed stability, partial, or complete response at three months. ‘Non-classified’ lesions were those with new or changed treatment during follow-up. Results: A cohort of 98 patients with 233 lesions was included over a 13-year period; 170 lesions were considered non-classified, and 41 and 22 lesions were classified as TP and pseudoprogression respectively. This resulted in a lesion- and patient-specific incidence for pseudoprogression of 9.4% and 17.3% respectively. Due to the large number of lesions that could not be classified, as is the case in clinical practice, the reported incidence in this study is likely an underestimation and can be seen as a ‘minimum’ incidence rate. Ten pseudoprogression (45.5%) and 13 (31.7%) TP lesions were previously irradiated. Pseudoprogression occurred at a median of 2.7 months after starting ICI therapy. The only clinical feature distinguishing patients with TP from pseudoprogression was that TP patients were more likely to need dexamethasone for neurological symptoms. Conclusions: Pseudoprogression has a lesion-specific incidence rate of at least 9.4% and occurs at a median of 2.7 months after starting ICI therapy. Severe neurological symptoms requiring dexamethasone may be a clinical feature typical for TP. Full article
(This article belongs to the Special Issue Feature Papers in the Section “Cancer Therapy” in 2025)
Show Figures

Figure 1

17 pages, 1361 KiB  
Review
Molecular Foundations of Neuroplasticity in Brain Tumours: From Microscopic Adaptation to Functional Reorganisation
by Lizeth Vinueza, Salvador Pineda and Jose E. Leon-Rojas
Int. J. Mol. Sci. 2025, 26(15), 7049; https://doi.org/10.3390/ijms26157049 - 22 Jul 2025
Viewed by 281
Abstract
Brain tumours challenge the structural and functional integrity of the brain, yet remarkable neuroplastic adaptations often preserve critical functions. This review synthesises the current knowledge of the molecular events underlying neuroplasticity in the context of tumoural growth, spanning from early genetic and protein [...] Read more.
Brain tumours challenge the structural and functional integrity of the brain, yet remarkable neuroplastic adaptations often preserve critical functions. This review synthesises the current knowledge of the molecular events underlying neuroplasticity in the context of tumoural growth, spanning from early genetic and protein alterations to macroscopic functional reorganisation. We discuss the roles of stress-regulated molecules, synaptic proteins, trophic factors, and morphological changes in driving adaptive responses. Furthermore, we bridge the gap between microscopic molecular events and large-scale network adaptations, emphasising clinical implications for glioma surgery and patient outcomes. Despite advances, knowledge gaps persist regarding the dynamics, predictors, and therapeutic modulation of plasticity, underscoring the need for future longitudinal and translational research. Understanding and leveraging these molecular mechanisms holds promise for improving functional recovery and quality of life in patients with brain tumours. Full article
(This article belongs to the Special Issue Brain Plasticity in Health and Disease)
Show Figures

Figure 1

24 pages, 1532 KiB  
Review
Polymeric Nanoparticle-Mediated Photodynamic Therapy: A Synergistic Approach for Glioblastoma Treatment
by Bandar Aldhubiab and Rashed M. Almuqbil
Pharmaceuticals 2025, 18(7), 1057; https://doi.org/10.3390/ph18071057 - 18 Jul 2025
Viewed by 443
Abstract
Glioblastoma is the most common and aggressive malignant primary brain tumour. Patients with glioblastoma have a median survival of only around 14.6 months after diagnosis, despite the availability of various conventional multimodal treatments including chemotherapy, radiation therapy, and surgery. Therefore, photodynamic therapy (PDT) [...] Read more.
Glioblastoma is the most common and aggressive malignant primary brain tumour. Patients with glioblastoma have a median survival of only around 14.6 months after diagnosis, despite the availability of various conventional multimodal treatments including chemotherapy, radiation therapy, and surgery. Therefore, photodynamic therapy (PDT) has emerged as an advanced, selective and more controlled therapeutic approach, which has minimal systemic toxicity and fewer side effects. PDT is a less invasive therapy that targets all cells or tissues that possess the photosensitizer (PS) itself, without affecting the surrounding healthy tissues. Polymeric NPs (PNPs) as carriers can improve the targeting ability and stability of PSs and co-deliver various anticancer agents to achieve combined cancer therapy. Because of their versatile tuneable features, these PNPs have the capacity to open tight junctions of the blood–brain barrier (BBB), easily transport drugs across the BBB, protect against enzymatic degradation, prolong the systemic circulation, and sustainably release the drug. Conjugated polymer NPs, poly(lactic-co-glycolic acid)-based NPs, lipid–polymer hybrid NPs, and polyethylene-glycolated PNPs have demonstrated great potential in PDT owing to their unique biocompatibility and optical properties. Although the combination of PDT and PNPs has great potential and can provide several benefits over conventional cancer therapies, there are several limitations that are hindering its translation into clinical use. This review aims to summarize the recent advances in the combined use of PNPs and PDT in the case of glioblastoma treatment. By evaluating various types of PDT and PNPs, this review emphasizes how these innovative approaches can play an important role in overcoming glioblastoma-associated critical challenges, including BBB and tumour heterogeneity. Furthermore, this review also discusses the challenges and future directions for PNPs and PDT, which provides insight into the potential solutions to various problems that are hindering their clinical translation in glioblastoma treatment. Full article
(This article belongs to the Special Issue Tumor Therapy and Drug Delivery)
Show Figures

Graphical abstract

18 pages, 2563 KiB  
Article
The Potential Anti-Cancer Effects of Polish Ethanolic Extract of Propolis and Quercetin on Glioma Cells Under Hypoxic Conditions
by Małgorzata Kłósek, Anna Kurek-Górecka, Radosław Balwierz, Grażyna Pietsz and Zenon P. Czuba
Molecules 2025, 30(14), 3008; https://doi.org/10.3390/molecules30143008 - 17 Jul 2025
Viewed by 649
Abstract
Tissue hypoxia is commonly observed in head cancers and contributes to both molecular and functional changes in tumour cells. It is known to stimulate erythropoiesis, angiogenesis, and metabolic alterations within tumour cells. Glioblastoma, a type of brain tumour, is characterized by rapid proliferation [...] Read more.
Tissue hypoxia is commonly observed in head cancers and contributes to both molecular and functional changes in tumour cells. It is known to stimulate erythropoiesis, angiogenesis, and metabolic alterations within tumour cells. Glioblastoma, a type of brain tumour, is characterized by rapid proliferation and aggressive growth. Recent studies have indicated that natural products may hold potential as components of cancer therapy. Among these, Polish propolis and its active compound, quercetin, have demonstrated promising anti-cancer properties. The aim of this study was to evaluate the concentrations of selected cytokines—specifically IL-6, IL-9, vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF-BB), interferon gamma-induced protein 10 (IP-10), and monocyte chemoattractant protein-1 (MCP-1)—produced by astrocytes of the CCF-STTG1 cell line. The cytotoxic effects of ethanolic extract of propolis (EEP) and quercetin were assessed using the MTT assay. Astrocytes were stimulated with lipopolysaccharide (LPS, 200 ng/mL) and/or IFN-α (100 U/mL), followed by treatment with EEP or quercetin (25–50 µg/mL) under hypoxic conditions for two hours. Cytokine concentrations were measured using the xMAP Luminex Multiplex Immunoassay and the Multiplex Bead-Based Cytokine Kit. Our study demonstrated that Polish propolis and its component quercetin modulate the tumour microenvironment in vitro, primarily by altering the levels of specific cytokines. The HCA analysis revealed that IL-6 and MCP-1 formed a distinct cluster at the highest linkage distance (approximately 100% of Dmax), suggesting that their expression patterns are significantly different from those of the other cytokines and that they are more similar to each other than to the rest. PCA analysis showed that EEP-PL (50 μg/mL) with IFN-α and EEP-PL (50 μg/mL) with LPS exert similar activities on cytokine secretion by astrocytes. Similar effects were demonstrated for EEP-PL 50 μg/mL + LPS + IFN-α, EEP-PL 25 μg/mL + IFN-α and EEP-PL 25 μg/mL + LPS + IFN-α. Our findings suggest that Polish propolis and quercetin may serve as promising natural agents to support the treatment of stage IV malignant astrocytoma. Nonetheless, further research is needed to confirm these results. Full article
Show Figures

Figure 1

20 pages, 1606 KiB  
Article
Brain Tumour Segmentation Using Choquet Integrals and Coalition Game
by Makhlouf Derdour, Mohammed El Bachir Yahiaoui, Moustafa Sadek Kahil, Mohamed Gasmi and Mohamed Chahine Ghanem
Information 2025, 16(7), 615; https://doi.org/10.3390/info16070615 - 17 Jul 2025
Viewed by 261
Abstract
Artificial Intelligence (AI) and computer-aided diagnosis (CAD) have revolutionised various aspects of modern life, particularly in the medical domain. These technologies enable efficient solutions for complex challenges, such as accurately segmenting brain tumour regions, which significantly aid medical professionals in monitoring and treating [...] Read more.
Artificial Intelligence (AI) and computer-aided diagnosis (CAD) have revolutionised various aspects of modern life, particularly in the medical domain. These technologies enable efficient solutions for complex challenges, such as accurately segmenting brain tumour regions, which significantly aid medical professionals in monitoring and treating patients. This research focuses on segmenting glioma brain tumour lesions in MRI images by analysing them at the pixel level. The aim is to develop a deep learning-based approach that enables ensemble learning to achieve precise and consistent segmentation of brain tumours. While many studies have explored ensemble learning techniques in this area, most rely on aggregation functions like the Weighted Arithmetic Mean (WAM) without accounting for the interdependencies between classifier subsets. To address this limitation, the Choquet integral is employed for ensemble learning, along with a novel evaluation framework for fuzzy measures. This framework integrates coalition game theory, information theory, and Lambda fuzzy approximation. Three distinct fuzzy measure sets are computed using different weighting strategies informed by these theories. Based on these measures, three Choquet integrals are calculated for segmenting different components of brain lesions, and their outputs are subsequently combined. The BraTS-2020 online validation dataset is used to validate the proposed approach. Results demonstrate superior performance compared with several recent methods, achieving Dice Similarity Coefficients of 0.896, 0.851, and 0.792 and 95% Hausdorff distances of 5.96 mm, 6.65 mm, and 20.74 mm for the whole tumour, tumour core, and enhancing tumour core, respectively. Full article
Show Figures

Figure 1

24 pages, 14721 KiB  
Article
Loss of 4.1B Drives PRMT3-Mediated Regulation of GBM Brain Tumour Stem Cell Growth
by Ravinder K. Bahia, Kyle Heemskerk, Samir Assaf, Orsolya Cseh, Xiaoguang Hao, Rozina Hassam, Panagiotis Prinos, H. Artee Luchman and Samuel Weiss
Int. J. Transl. Med. 2025, 5(3), 29; https://doi.org/10.3390/ijtm5030029 - 7 Jul 2025
Viewed by 442
Abstract
Background: Protein arginine methyltransferase 3 (PRMT3), a type I family PRMT, regulates the activity of downstream substrates by catalyzing the asymmetric dimethylation of arginine residues. While PRMT3 activity has been reported to be deregulated in many cancers, including glioblastoma (GBM), the underlying signalling [...] Read more.
Background: Protein arginine methyltransferase 3 (PRMT3), a type I family PRMT, regulates the activity of downstream substrates by catalyzing the asymmetric dimethylation of arginine residues. While PRMT3 activity has been reported to be deregulated in many cancers, including glioblastoma (GBM), the underlying signalling mechanisms that contribute to disease progression are largely unknown. Methods: We tested the efficacy of a PRMT3 chemical probe, SGC707, in a cohort of GBM patient-derived primary and recurrent brain tumour stem cell (BTSC) lines. RNA-sequencing, CRISPR-cas9 knockout, and inducible overexpression methods were used to investigate the molecular mechanisms regulated by the aberrant activity of PRMT3 in different BTSC lines. Results: We show that expression of the tumour suppressor protein 4.1B, a negative regulator of PRMT3, predicts the response of GBM BTSCs to the PRMT3 chemical probe, SGC707. Furthermore, PRMT3 modulates the stability and subcellular localization of the downstream effector, UHRF1, a member of the DNA methylation complex. These findings suggest that UHRF1 and DNMT1 may suppress the expression of 4.1B through the increased promoter methylation of EPB4.1L3. Intriguingly, the inducible overexpression of EPB4.1L3 in the BT248EPB4.1L3low BTSC line mimicked the effects of the pharmacologic and genetic inhibition of PRMT3. In contrast, knockout of EPB4.1L3 in BT143EPB4.1L3high cells reduced the interactions between PRMT3 and 4.1B proteins, resulting in increased sensitivity of knockout cells to SGC707 treatment. Conclusions: These findings show that 4.1B, PRMT3, and UHRF1/DNMT1 function together to promote BTSC growth. Thus, targeting PRMT3 or UHRF1/DNMT1, especially in tumours with low endogenous 4.1B protein, may have high therapeutic relevance. Full article
Show Figures

Figure 1

22 pages, 1990 KiB  
Article
Circadian-Tuned Peptide Drug/Gene Co-Delivery Nanocomplexes to Enhance Glioblastoma Targeting and Transfection
by Ana R. Neves, Eric Vivès, Prisca Boisguérin, Telma Quintela and Diana Costa
Int. J. Mol. Sci. 2025, 26(13), 6130; https://doi.org/10.3390/ijms26136130 - 26 Jun 2025
Viewed by 584
Abstract
Glioblastoma is the most prevalent and aggressive form of brain malignancy. Actual treatments face several challenges due to its high aggressiveness and poor prognosis. The chemotherapeutic agent temozolomide (TMZ) has limited therapeutic efficacy, and mutations in the tumour protein p53 gene (TP53 [...] Read more.
Glioblastoma is the most prevalent and aggressive form of brain malignancy. Actual treatments face several challenges due to its high aggressiveness and poor prognosis. The chemotherapeutic agent temozolomide (TMZ) has limited therapeutic efficacy, and mutations in the tumour protein p53 gene (TP53) have been associated with treatment resistance. Thus, this study aimed to explore an innovative therapeutic strategy to enhance treatment efficacy of GBM. Previously, our team had developed a WRAP5 cell-penetrating peptide (CPP) functionalized with a transferrin receptor ligand (Tf) for the targeted delivery of TMZ and a p53-encoding plasmid to glioma cells. Our research had elucidated the circadian oscillations of the clock genes in the U87 glioma cells by employing two different computational models and observed that T16 and T8 time points revealed the highest circadian activity for Bmal1 and Per2 genes, respectively. Similar analysis was conducted for the transferrin receptor, which revealed that T7 and T8 were the key time points for its expression. A confocal microscopy study indicated the highest intracellular uptake of complexes and p53 mRNA expression at T8, the time point with the highest Per2 and transferrin receptor expression. Following mRNA analysis, the evaluation of p53 levels confirmed transcriptional changes at the protein level, and that T16 appears to be a favourable time point for enhancing therapeutic efficacy in U87 glioblastoma cells. These findings suggested that synchronizing the complexes’ administration with the biological clock of GBM cells may significantly improve glioblastoma therapeutics. Full article
(This article belongs to the Special Issue The Importance of Molecular Circadian Rhythms in Health and Disease)
Show Figures

Graphical abstract

9 pages, 757 KiB  
Communication
Sugar-Linked Diethyldithiocarbamate Derivatives: A Novel Class of Anticancer Agents
by Mohammad Najlah, Niamh McCallum, Ana Maria Pereira, Dan Alves, Niusha Ansari-Fard, Sahrish Rehmani and Ayşe Kaya
Int. J. Mol. Sci. 2025, 26(12), 5589; https://doi.org/10.3390/ijms26125589 - 11 Jun 2025
Viewed by 359
Abstract
Disulfiram (DSF), a well-known anti-alcoholism drug, exhibits potent anticancer activity via its metabolite, diethyldithiocarbamate (DDC), which forms a cytotoxic copper complex that selectively targets cancer stem cells. However, its clinical utility is limited by poor solubility and rapid plasma metabolism. This study explores [...] Read more.
Disulfiram (DSF), a well-known anti-alcoholism drug, exhibits potent anticancer activity via its metabolite, diethyldithiocarbamate (DDC), which forms a cytotoxic copper complex that selectively targets cancer stem cells. However, its clinical utility is limited by poor solubility and rapid plasma metabolism. This study explores saccharide-linked DDCs as novel prodrugs designed to enhance stability, solubility, and tumour-selective activation. These compounds feature thioglycosidic bonds that shield the DDC moiety from premature degradation while retaining its metal-chelating function to form the active copper(II)bis(N,N-diethyldithiocarbamate) (Cu(DDC)2) complex. The synthesised derivatives were characterised and evaluated for serum stability and in vitro cytotoxicity across several cancer cell lines, including colorectal, breast, lung, and brain cancers. Copper-complexed saccharide-DDC prodrugs demonstrated remarkable cytotoxicity, with improved biostability and solubility profiles. These findings highlight the potential of saccharide-linked DDCs as stable, copper-activated prodrugs for cancer therapy. Further in vivo studies are warranted to validate their pharmacokinetics and clinical relevance. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Figure 1

21 pages, 4793 KiB  
Article
Deep Learning for Glioblastoma Multiforme Detection from MRI: A Statistical Analysis for Demographic Bias
by Kebin Contreras, Julio Gutierrez-Rengifo, Oscar Casanova-Carvajal, Angel Luis Alvarez, Patricia E. Vélez-Varela and Ana Lorena Urbano-Bojorge
Appl. Sci. 2025, 15(11), 6274; https://doi.org/10.3390/app15116274 - 3 Jun 2025
Viewed by 640
Abstract
Glioblastoma, IDH-wildtype (GBM), is the most aggressive and complex brain tumour classified by the World Health Organization (WHO), characterised by high mortality rates and diagnostic limitations inherent to invasive conventional procedures. Early detection is essential for improving patient outcomes, underscoring the need for [...] Read more.
Glioblastoma, IDH-wildtype (GBM), is the most aggressive and complex brain tumour classified by the World Health Organization (WHO), characterised by high mortality rates and diagnostic limitations inherent to invasive conventional procedures. Early detection is essential for improving patient outcomes, underscoring the need for non-invasive diagnostic tools. This study presents a convolutional neural network (CNN) specifically optimised for GBM detection from T1-weighted magnetic resonance imaging (MRI), with systematic evaluations of layer depth, activation functions, and hyperparameters. The model was trained on the RSNA-MICCAI data set and externally validated on the Erasmus Glioma Database (EGD), which includes gliomas of various grades and preserves cranial structures, unlike the skull-stripped RSNA-MICCAI images. This morphological discrepancy demonstrates the generalisation capacity of the model across anatomical and acquisition differences, achieving an F1-score of 0.88. Furthermore, statistical tests, such as Shapiro–Wilk, Mann–Whitney U, and Chi-square, confirmed the absence of demographic bias in model predictions, based on p-values, confidence intervals, and statistical power analyses supporting its demographic fairness. The proposed model achieved an area under the curve–receiver operating characteristic (AUC-ROC) of 0.63 on the RSNA-MICCAI test set, surpassing all prior results submitted to the BraTS 2021 challenge, and establishing a reliable and generalisable approach for non-invasive GBM detection. Full article
(This article belongs to the Special Issue Convolutional Neural Networks and Computer Vision)
Show Figures

Figure 1

30 pages, 6234 KiB  
Article
Molecular Effects of Cornelian Cherry Fruit (Cornus mas L.) Extract on Sleep Deprivation-Induced Oxidative Stress, Cytokine Dysregulation, and Behavioural Changes in Wistar Rats
by Vlad Sever Neculicioiu, Ioana Colosi, Alexandra Sevastre-Berghian, Dan Alexandru Toc, Horațiu Alexandru Colosi, Luminita David, Mara Muntean, Remus Moldovan, Ana-Maria Vlase, Vlad Alexandru Toma, Carmen Costache, Şoimiţa Mihaela Suciu and Simona Clichici
Curr. Issues Mol. Biol. 2025, 47(6), 399; https://doi.org/10.3390/cimb47060399 - 28 May 2025
Viewed by 627
Abstract
Sleep deprivation (SD) induces significant neurobiological changes, including oxidative stress, neuroinflammation, and behavioural impairments. This study was designed as a proof of concept to assess the potential for modulating the effects of SD through a short-term seven-day administration of Cornus mas (C. [...] Read more.
Sleep deprivation (SD) induces significant neurobiological changes, including oxidative stress, neuroinflammation, and behavioural impairments. This study was designed as a proof of concept to assess the potential for modulating the effects of SD through a short-term seven-day administration of Cornus mas (C. mas) in a rapid eye movement (REM) SD rodent paradigm. Adult male Wistar rats were randomised in four groups (n = 7): control, C. mas (CM), sleep deprivation (SD), and sleep deprivation with C. mas (SD + CM). Behaviourally, SD induced hyperactivity and hyperlocomotion. SD determined histological alterations in the prefrontal cortex and corpus callosum myelin coupled with ultrastructural mitochondrial and cellular abnormalities in the prefrontal cortex, hippocampus, and pineal gland. Despite evidence of systemic oxidative stress coupled with decreased serum GABA and BDNF following SD, no significant changes were observed in redox markers or inflammatory cytokine levels (TNF-α, IL-1β) within the prefrontal cortex or hippocampus. C. mas extract has shown an overall modest modulatory action, mainly evidenced on behavioural, histological, and ultrastructural parameters. Taken together, these findings highlight behavioural changes and region-specific molecular and structural abnormalities following prolonged REM SD in rats. Full article
Show Figures

Figure 1

21 pages, 594 KiB  
Review
Spatial Mapping of Glioblastoma Infiltration: Diffusion Tensor Imaging-Based Radiomics and Connectomics in Recurrence Prediction
by Kevin Jang and Michael Back
Brain Sci. 2025, 15(6), 576; https://doi.org/10.3390/brainsci15060576 - 27 May 2025
Viewed by 875
Abstract
Glioblastoma (GBM) often exhibits distinct anatomical patterns of relapse after radiotherapy. Tumour cell migration along myelinated white matter tracts is a key driver of disease progression. The failure of conventional imaging to capture subclinical infiltration has driven interest in advanced imaging biomarkers capable [...] Read more.
Glioblastoma (GBM) often exhibits distinct anatomical patterns of relapse after radiotherapy. Tumour cell migration along myelinated white matter tracts is a key driver of disease progression. The failure of conventional imaging to capture subclinical infiltration has driven interest in advanced imaging biomarkers capable of quantifying tumour–brain interactions. Diffusion tensor imaging (DTI), radiomics, and connectomics represent a triad of innovative, non-invasive approaches that map white matter architecture, predict recurrence risk, and inform biologically guided treatment strategies. This review examines the biological rationale and clinical applications of DTI-based metrics, radiomic signatures, and tractography-informed connectomics in GBM. We discuss the integration of these modalities into machine learning frameworks and radiotherapy/surgical planning, supported by landmark studies and multi-institutional data. The implications for personalised neuro-oncology are profound, marking a shift towards risk-adaptive, tract-aware treatment strategies that may improve local control and preserve neurocognitive function. Full article
(This article belongs to the Special Issue Editorial Board Collection Series: Advances in Neuro-Oncology)
Show Figures

Figure 1

22 pages, 3829 KiB  
Article
Brain Tumour Segmentation and Grading Using Local and Global Context-Aggregated Attention Network Architecture
by Ahmed Abdulhakim Al-Absi, Rui Fu, Nadhem Ebrahim, Mohammed Abdulhakim Al-Absi and Dae-Ki Kang
Bioengineering 2025, 12(5), 552; https://doi.org/10.3390/bioengineering12050552 - 21 May 2025
Viewed by 631
Abstract
Brain tumours (BTs) are among the most dangerous and life-threatening cancers in humans of all ages, and the early detection of BTs can make a huge difference to their treatment. However, grade recognition is a challenging issue for radiologists involved in automated diagnosis [...] Read more.
Brain tumours (BTs) are among the most dangerous and life-threatening cancers in humans of all ages, and the early detection of BTs can make a huge difference to their treatment. However, grade recognition is a challenging issue for radiologists involved in automated diagnosis and healthcare monitoring. Recent research has been motivated by the search for deep learning-based mechanisms for segmentation and grading to assist radiologists in diagnostic analysis. Segmentation refers to the identification and delineation of tumour regions in medical images, while classification classifies based on tumour characteristics, such as the size, location and enhancement pattern. The main aim of this research is to design and develop an intelligent model that can detect and grade tumours more effectively. This research develops an aggregated architecture called LGCNet, which combines a local context attention network and a global context attention network. LGCNet makes use of information extracted through the task, dimension and scale. Specifically, a global context attention network is developed for capturing multiple-scale features, and a local context attention network is designed for specific tasks. Thereafter, both networks are aggregated, and the learning network is designed to balance all the tasks by combining the loss functions of the classification and segmentation. The main advantage of LGCNet is its dedicated network for a specific task. The proposed model is evaluated by considering the BraTS2019 dataset with different metrics, such as the Dice score, sensitivity, specificity and Hausdorff score. Comparative analysis with the existing model shows marginal improvement and provides scope for further research into BT segmentation and classification. The scope of this study focuses on the BraTS2019 dataset, with future work aiming to extend the applicability of the model to different clinical and imaging environments. Full article
(This article belongs to the Section Biosignal Processing)
Show Figures

Figure 1

21 pages, 10416 KiB  
Article
Combinational Radiotherapies Improve Brain Cancer Treatment at High Dose Rates In Vitro
by Michael Valceski, Elette Engels, Sarah Vogel, Jason Paino, Dylan Potter, Carolyn Hollis, Abass Khochaiche, Micah Barnes, Alice O’Keefe, Matthew Cameron, Kiarn Roughley, Anatoly Rosenfeld, Michael Lerch, Stéphanie Corde and Moeava Tehei
Cancers 2025, 17(10), 1713; https://doi.org/10.3390/cancers17101713 - 20 May 2025
Viewed by 3234
Abstract
Background/objectives: Brain cancer remains difficult to treat, with survival statistics stagnant for decades. The resistance of glioblastoma brain tumours can greatly challenge the effectiveness of conventional cancer radiotherapy. However, high dose rate radiotherapy has unique effects that allow for normal tissue sparing whilst [...] Read more.
Background/objectives: Brain cancer remains difficult to treat, with survival statistics stagnant for decades. The resistance of glioblastoma brain tumours can greatly challenge the effectiveness of conventional cancer radiotherapy. However, high dose rate radiotherapy has unique effects that allow for normal tissue sparing whilst maintaining tumour control. The addition of targeted radiosensitisers, such as the chemotherapeutic drug methotrexate (MTX) or the high-Z halogenated pyrimidine drug iododeoxyuridine (IUdR), can improve radiotherapy outcomes. Combining these radiosensitiser agents with ultra-high dose rate (UHDR) synchrotron X-rays can bear synergistic effects to enhance the efficacy of these multi-modal UHDR therapies, providing a means to overcome the radioresistance of brain cancer. Methods: Here, we use controlled in vitro assays following treatment, including a clonogenic assay to determine long-term cell survival and γH2AX immunofluorescent confocal microscopy to quantify double-strand DNA breaks (DSBs). Results: We find significant enhancement for highly synergistic combinations of IUdR+MTX with synchrotron X-rays. Cell survival results demonstrate 5.4 times increased 9L gliosarcoma cell killing when these agents are combined with UHDR synchrotron X-rays compared with conventional X-rays alone at the same 5 Gy dose. The underlying mechanisms are unveiled using γH2AX imaging and reveal significant increases in DSBs and dying cells following exposure to UHDR radiation. Conclusions: Our results demonstrate that highly synergistic combination treatments using UHDR synchrotron radiation can yield significantly improved brain cancer killing compared with conventional radiotherapy. We anticipate that these additive, multi-modal combination therapies will provide options for more targeted and effective use of radiotherapies for the future treatment of brain cancer. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Graphical abstract

30 pages, 2663 KiB  
Review
IGFBP-2 and IGF-II: Key Components of the Neural Stem Cell Niche? Implications for Glioblastoma Pathogenesis
by Abigail J. Harland and Claire M. Perks
Int. J. Mol. Sci. 2025, 26(10), 4749; https://doi.org/10.3390/ijms26104749 - 15 May 2025
Viewed by 1113
Abstract
Glioblastoma is a fatal and aggressive cancer with no cure. It is becoming increasingly clear that glioblastoma initiation is a result of adult neural stem cell (NSC) transformation—most likely those within the subventricular zone (SVZ). Indeed, transcriptomic analysis indicates that glioblastomas are reminiscent [...] Read more.
Glioblastoma is a fatal and aggressive cancer with no cure. It is becoming increasingly clear that glioblastoma initiation is a result of adult neural stem cell (NSC) transformation—most likely those within the subventricular zone (SVZ). Indeed, transcriptomic analysis indicates that glioblastomas are reminiscent of a neurodevelopmental hierarchy, in which neural stem and progenitor markers are widely expressed by tumour stem-like cells. However, NSC fates and the cues that drive them are poorly understood. Studying the crosstalk within NSC niches may better inform our understanding of glioblastoma initiation and development. Insulin-like growth factor binding protein 2 (IGFBP-2) has a well-established prognostic role in glioblastoma, and cell-based mechanistic studies show the independent activation of downstream oncogenic pathways. However, IGFBP-2 is more commonly recognised as a modulator of insulin-like growth factors (IGFs) for receptor tyrosine kinase signal propagation or attenuation. In the adult human brain, both IGFBP-2 and IGF-II expression are retained in the choroid plexus (ChP) and secreted into the cerebral spinal fluid (CSF). Moreover, secretion by closely associated cells and NSCs themselves position IGFBP-2 and IGF-II as interesting factors within the NSC niche. In this review, we will highlight the experimental findings that show IGFBP-2 and IGF-II influence NSC behaviour. Moreover, we will link this to glioblastoma biology and demonstrate the requirement for further analysis of these factors in glioma stem cells (GSCs). Full article
(This article belongs to the Special Issue The Role of the IGF Axis in Disease, 4th Edition)
Show Figures

Figure 1

Back to TopTop