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

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (186)

Search Parameters:
Keywords = brain tumor initiating cells

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 13869 KiB  
Article
Spatial Omics Profiling of Treatment-Naïve Lung Adenocarcinoma with Brain Metastasis as the Initial Presentation
by Seoyeon Gwon, Inju Cho, Jieun Lee, Seung Yun Lee, Kyue-Hee Choi and Tae-Jung Kim
Cancers 2025, 17(15), 2529; https://doi.org/10.3390/cancers17152529 - 31 Jul 2025
Viewed by 87
Abstract
Background/Objectives: Brain metastasis (BM) is a common and often early manifestation in lung adenocarcinoma (LUAD), yet its tumor microenvironment remains poorly defined at the time of initial diagnosis. This study aims to characterize early immune microenvironmental alterations in synchronous BM using spatial proteomic [...] Read more.
Background/Objectives: Brain metastasis (BM) is a common and often early manifestation in lung adenocarcinoma (LUAD), yet its tumor microenvironment remains poorly defined at the time of initial diagnosis. This study aims to characterize early immune microenvironmental alterations in synchronous BM using spatial proteomic profiling. Methods: We performed digital spatial proteomic profiling using the NanoString GeoMx platform on formalin-fixed paraffin-embedded tissues from five treatment-naïve LUAD patients in whom BM was the initial presenting lesion. Paired primary lung and brain metastatic samples were analyzed across tumor and stromal compartments using 68 immune- and tumor-related protein markers. Results: Spatial profiling revealed distinct expression patterns between primary tumors and brain metastases. Immune regulatory proteins—including IDO-1, PD-1, PD-L1, STAT3, PTEN, and CD44—were significantly reduced in brain metastases (p < 0.01), whereas pS6, a marker of activation-induced T-cell death, was significantly upregulated (p < 0.01). These alterations were observed in both tumor and stromal regions, suggesting a more immunosuppressive and apoptotic microenvironment in brain lesions. Conclusions: This study provides one of the first spatially resolved proteomic characterizations of synchronous BM at initial LUAD diagnosis. Our findings highlight early immune escape mechanisms and suggest the need for site-specific immunotherapeutic strategies in patients with brain metastasis. Full article
(This article belongs to the Special Issue Lung Cancer Proteogenomics: New Era, New Insights)
Show Figures

Figure 1

18 pages, 8645 KiB  
Article
CIC-Rearranged Sarcoma: A Clinical and Pathological Study of a Peculiar Entity
by Ward Maaita, Nabil Hasasna, Sameer Yaser, Yacob Saleh, Ramiz Abu-Hijlih, Wafa Asha, Hadeel Halalsheh, Samer Abdel Al, Maysa Al-Hussaini and Omar Jaber
Diagnostics 2025, 15(14), 1758; https://doi.org/10.3390/diagnostics15141758 - 11 Jul 2025
Viewed by 460
Abstract
Background: CIC-rearranged sarcoma is a rare and aggressive type of undifferentiated round cell tumor characterized by CIC gene fusion, most commonly CIC::DUX4. This study presents a series of eleven cases, highlighting their clinicopathological features. Methods: Pathology records (2019 to 2024) [...] Read more.
Background: CIC-rearranged sarcoma is a rare and aggressive type of undifferentiated round cell tumor characterized by CIC gene fusion, most commonly CIC::DUX4. This study presents a series of eleven cases, highlighting their clinicopathological features. Methods: Pathology records (2019 to 2024) were searched using “sarcoma with CIC”, identifying eleven cases, of which seven referred cases were initially misdiagnosed. Pathological and clinical analysis was conducted. Treatment was dictated upon multidisciplinary panel discussion based on tumor stage. Follow-up data (1–25 months) was available for all patients. Results: The cohort included six males and five females, with a median age of 43 years (range;14–53), with nine in soft tissue and two in bone. Tumor size ranged from 3.5 cm to 20.0 cm (mean: 9.8 cm). Most cases showed sheets of undifferentiated round- to oval-shaped cells. Two cases showed an Ewing-like pattern, and one case showed spindle cells in a fibrotic stroma transitioning to epithelioid cells. Necrosis was present in nine cases, and mitotic count ranged from 2 to 38/ 10HPFs (mean = 14.2). CD99 was positive in (10/11) cases and WT-1 in (6/9). NKX2.2, S100, and MDM2 were positive in rare cases. CIC::DUX4 fusion was detected in four cases. FISH for CIC gene rearrangement was positive in seven cases, two of them confirmed by methylation analysis. Metastasis at diagnosis was common (n = 8), primarily in the lungs, with later metastasis to the brain and bone. At time of final analysis, eight patients died within a median of 10 months (range: 1–19 months), while three were alive, two with stable disease (for a period of 6 and 25 months) and one with progression after 10 months. Significant correlation was seen between overall survival and the presence of metastasis at diagnosis (p value = 0.03). Conclusions: CIC-rearranged sarcomas are rare, high-grade tumors with predilection for soft tissue. Misdiagnosis is frequent, necessitating molecular confirmation. These tumors are treatment-resistant, often present with lung metastasis, and carry a poor prognosis, especially with initial metastasis. Full article
(This article belongs to the Section Pathology and Molecular Diagnostics)
Show Figures

Figure 1

16 pages, 1236 KiB  
Communication
Chemoradiation-Altered Micromilieu of Glioblastoma Cells Particularly Impacts M1-like Macrophage Activation
by Mona Shojaei, Benjamin Frey, Florian Putz, Rainer Fietkau, Udo S. Gaipl and Anja Derer
Int. J. Mol. Sci. 2025, 26(14), 6574; https://doi.org/10.3390/ijms26146574 - 8 Jul 2025
Viewed by 401
Abstract
Glioblastoma is a highly aggressive brain tumor with an overall poor prognosis due to its immunosuppressive tumor microenvironment (TME). Microglia and tumor-associated macrophages (TAMs) with pro-tumorigenic properties are dominant populations of immune cells in the glioblastoma TME. To date, several studies targeting TAMs [...] Read more.
Glioblastoma is a highly aggressive brain tumor with an overall poor prognosis due to its immunosuppressive tumor microenvironment (TME). Microglia and tumor-associated macrophages (TAMs) with pro-tumorigenic properties are dominant populations of immune cells in the glioblastoma TME. To date, several studies targeting TAMs to fight tumor progression in different tumor entities have been initiated. However, the impact of standard therapy schemes of glioblastoma cells on macrophage polarization, activation, and phagocytosis remains controversial. The same applies to the relevance of PD-1/PD-L1 blockade in the interaction between macrophages and tumor cells. Our study, therefore, investigated patient-oriented treatment of GLIOBLASTOMA by examining the phagocytic capacity of polarized M1- and M2-like macrophages using GL261-luc2 tumor cells as a preclinical model system. Additionally, we analyzed the expression of activation and immune checkpoint markers on these macrophage subtypes following contact with tumor cells and their microenvironment. These factors were also determined after PD-1 blockade was initiated. The analyses revealed that the immunoregulatory M2-like macrophages generally exhibited a higher phagocytosis rate than the pro-inflammatory M1-like macrophages; however, this was not influenced by the pretreatment of glioblastoma cells with chemo- or radiotherapy. This could not be improved by blocking the PD-1 receptor. Furthermore, there were no modulations in the expression of differentiation, activation, or immune checkpoint molecules of M1- and M2-like macrophages after cell-to-cell contact with glioblastoma cells. But the medium conditioned by tumor cells strongly altered M1-like macrophages toward a more activated state, whereas M2-like cells were only mildly influenced. This was further enhanced by tumor cell treatment, with the most prominent effect after irradiation. These results suggest that conventional GLIOBLASTOMA tumor cell treatment affects the immunogenic status of macrophage subtypes, which is relevant for enhancing the anti-tumor immune response in brain tumors. Full article
(This article belongs to the Special Issue The Role of Macrophages in Cancers)
Show Figures

Figure 1

37 pages, 1459 KiB  
Review
Current Landscape of Preclinical Models for Pediatric Gliomas: Clinical Implications and Future Directions
by Syed M. Faisal, Monika Yadav, Garrett R. Gibson, Adora T. Klinestiver, Ryan M. Sorenson, Evan Cantor, Maria Ghishan, John R. Prensner, Andrea T. Franson, Kevin F. Ginn, Carl Koschmann and Viveka Nand Yadav
Cancers 2025, 17(13), 2221; https://doi.org/10.3390/cancers17132221 - 2 Jul 2025
Viewed by 1266
Abstract
Pediatric high-grade gliomas (pHGGs), particularly diffuse midline gliomas (DMGs), are among the most lethal brain tumors due to poor survival and resistance to therapies. DMGs possess a distinct genetic profile, primarily driven by hallmark mutations such as H3K27M, ACVR1, and PDGFRA mutations/amplifications and [...] Read more.
Pediatric high-grade gliomas (pHGGs), particularly diffuse midline gliomas (DMGs), are among the most lethal brain tumors due to poor survival and resistance to therapies. DMGs possess a distinct genetic profile, primarily driven by hallmark mutations such as H3K27M, ACVR1, and PDGFRA mutations/amplifications and TP53 inactivation, all of which contribute to tumor biology and therapeutic resistance. Developing physiologically relevant preclinical models that replicate both tumor biology and the tumor microenvironment (TME) is critical for advancing effective treatments. This review highlights recent progress in in vitro, ex vivo, and in vivo models, including patient-derived brain organoids, genetically engineered mouse models (GEMMs), and region-specific midline organoids incorporating SHH, BMP, and FGF2/8/19 signaling to model pontine gliomas. Key genetic alterations can now be introduced using lipofectamine-mediated transfection, PiggyBac plasmid systems, and CRISPR-Cas9, allowing the precise study of tumor initiation, progression, and therapy resistance. These models enable the investigation of TME interactions, including immune responses, neuronal infiltration, and therapeutic vulnerabilities. Future advancements involve developing immune-competent organoids, integrating vascularized networks, and applying multi-omics platforms like single-cell RNA sequencing and spatial transcriptomics to dissect tumor heterogeneity and lineage-specific vulnerabilities. These innovative approaches aim to enhance drug screening, identify new therapeutic targets, and accelerate personalized treatments for pediatric gliomas. Full article
Show Figures

Figure 1

32 pages, 5511 KiB  
Article
Development of Carbohydrate Polyelectrolyte Nanoparticles for Use in Drug Delivery Systems that Cross the Blood–Brain Barrier to Treat Brain Tumors
by Vladimir E. Silant’ev, Mikhail E. Shmelev, Andrei S. Belousov, Fedor O. Trukhin, Nadezhda E. Struppul, Aleksandra A. Patlay, Anna K. Kravchenko, Sergey P. Shchava and Vadim V. Kumeiko
Polymers 2025, 17(12), 1690; https://doi.org/10.3390/polym17121690 - 18 Jun 2025
Viewed by 487
Abstract
The low effectiveness of various brain cancer treatment methods is due to a number of significant challenges. Most of them are unable to penetrate the blood–brain barrier (BBB) when drugs are administered systemically through the bloodstream. Nanoscale particles play a special role among [...] Read more.
The low effectiveness of various brain cancer treatment methods is due to a number of significant challenges. Most of them are unable to penetrate the blood–brain barrier (BBB) when drugs are administered systemically through the bloodstream. Nanoscale particles play a special role among materials capable of binding drug molecules and successfully crossing the BBB. Biopolymeric nanoparticles (NPs) demonstrate excellent biocompatibility and have the remarkable ability to modify the environment surrounding tumor cells, thereby potentially improving cellular uptake of delivery agents. In our research, nanoscale polyelectrolyte complexes (PECs) ranging in size from 56 to 209 nm were synthesized by ionic interaction of the oppositely charged polysaccharides pectin and chitosan. The structural characteristics of these complexes were carefully characterized by infrared (FTIR) and Raman spectroscopy. The immobilization efficiency of antitumor drugs was comprehensively evaluated using UV spectrophotometry. The cytotoxicity of the NPs was evaluated in the U87-MG cell line. The preliminary data indicate a significant decrease in the metabolic activity of these tumor cells. Important details on the interaction of the NPs with an endothelial layer structurally similar to the BBB were obtained by simulating the BBB using a model based on human blood vessels. Our studies allowed us to establish a significant correlation between the kinetic parameters of drug immobilization and the ratio of biopolymer concentrations in the initial compositions, which provides valuable information for future optimization of drug delivery system design. Full article
(This article belongs to the Special Issue Advanced Polymeric Biomaterials for Drug Delivery Applications)
Show Figures

Figure 1

16 pages, 3480 KiB  
Case Report
Navigating Rarity: Pathological Challenges and Diagnostic Ambiguity in Rare Gliomas—A Case Series with a Focus on Personalized Treatment and Quality of Life
by Nadja Grübel, Anika Wickert, Felix Sahm, Bernd Schmitz, Anja Osterloh, Rebecca Kassubek, Ralph König, Christian Rainer Wirtz, Jens Engelke, Andrej Pala and Mona Laible
Onco 2025, 5(2), 28; https://doi.org/10.3390/onco5020028 - 10 Jun 2025
Viewed by 739
Abstract
Gliomas are incurable, heterogeneous brain tumors, with rare forms often constituting diagnostic and treatment challenges. Molecular diagnostics, mainly implemented through the World Health Organization (WHO) 2021 guidelines, have refined the classification, but highlight difficulties in diagnosing rare gliomas remain. This case series analyzes [...] Read more.
Gliomas are incurable, heterogeneous brain tumors, with rare forms often constituting diagnostic and treatment challenges. Molecular diagnostics, mainly implemented through the World Health Organization (WHO) 2021 guidelines, have refined the classification, but highlight difficulties in diagnosing rare gliomas remain. This case series analyzes four patients with rare gliomas treated at the University Hospital, Ulm, between 2002 and 2024. Patients were selected based on unique histopathological features and long-term clinical follow-up. Clinical records, imaging, and histological data were reviewed. Molecular diagnostics followed WHO 2021 guidelines. Quality of life was assessed using standardized tools including the EQ-5D-5L, EQ VAS, the Distress Thermometer, and the Montreal Cognitive Assessment (MoCA). In the first case, a 51-year-old male’s diagnosis evolved from pleomorphic xanthoastrocytoma to a high-grade glioma with pleomorphic and pseudopapillary features, later identified as a neuroepithelial tumor with a PATZ1 fusion over 12 years. Despite multiple recurrences, extensive surgical interventions led to excellent outcomes. The second case involved a young female with long-term survival of astroblastoma, demonstrating significant improvements in both longevity and quality of life through personalized care. The third case involved a patient with oligodendroglioma, later transforming into glioblastoma, emphasizing the importance of continuous diagnostic reevaluation and adaptive treatment strategies, contributing to prolonged survival and quality of life improvements. Remarkably, the patient has achieved over 20 years of survival, including 10 years of being both therapy- and progression-free. The fourth case presents a young woman with neurofibromatosis type 1, initially misdiagnosed with glioblastoma based on histopathological findings. Subsequent molecular diagnostics revealed a subependymal giant cell astrocytoma-like astrocytoma, highlighting the critical role of early advanced diagnostic techniques. These cases underscore the importance of precise molecular diagnostics, individualized treatments, and ongoing diagnostic reevaluation to optimize outcomes. They also address the psychological impact of evolving diagnoses, stressing the need for comprehensive patient support. Even in complex cases, extensive surgical interventions can yield favorable results, reinforcing the value of adaptive, multidisciplinary strategies based on evolving tumor characteristics. Full article
Show Figures

Figure 1

25 pages, 2883 KiB  
Article
Metabolic Reprogramming Triggered by Fluoride in U-87 Glioblastoma Cells: Implications for Tumor Progression?
by Wojciech Żwierełło, Agnieszka Maruszewska, Marta Skórka-Majewicz, Agata Wszołek and Izabela Gutowska
Cells 2025, 14(11), 800; https://doi.org/10.3390/cells14110800 - 29 May 2025
Viewed by 503
Abstract
Chronic inflammation is a hallmark of brain tumors, especially gliomas, which exhibit elevated levels of pro-inflammatory mediators within the tumor and its microenvironment. Metabolic disturbances triggered by fluoride as a pro-oxidative agent in glioma cells, known for their high aggressiveness and resistance to [...] Read more.
Chronic inflammation is a hallmark of brain tumors, especially gliomas, which exhibit elevated levels of pro-inflammatory mediators within the tumor and its microenvironment. Metabolic disturbances triggered by fluoride as a pro-oxidative agent in glioma cells, known for their high aggressiveness and resistance to therapy—remain poorly understood. Therefore, investigating the impact of physiologically elevated fluoride concentrations on oxidative stress and pro-inflammatory responses in glioma cells represents a relevant and timely research objective. Methods: U-87 human glioblastoma cells were subjected to short-term and long-term exposure to physiologically high concentrations of NaF (0.1–10 µM). Both the cells and the culture medium were analyzed. We assessed levels of reactive oxygen species (ROS), antioxidant defenses, and a panel of cytokines and chemokines. Results: Our results demonstrated that oxidative stress and inflammatory conditions in U-87 cells varied with fluoride concentration and exposure time. This led to an increase in ROS levels and key pro-inflammatory cytokines, including IL-6 and TNF-α. Conclusions: Fluoride compounds can generate ROS and disrupt the antioxidant defense system in U-87 human glioblastoma cells, leading to the initiation and progression of inflammatory states. Furthermore, prolonged exposure to NaF may induce adaptive mechanisms in U-87 cells. Full article
(This article belongs to the Special Issue Metabolic Hallmarks in Cancer)
Show Figures

Figure 1

17 pages, 17046 KiB  
Article
Olig1/2 Drive Astrocytic Glioblastoma Proliferation Through Transcriptional Co-Regulation of Various Cyclins
by Yu Tian, Ziwu Wang, Mengge Sun, Jialin Li, Wenhui Zheng, Feihong Yang and Zhuangzhi Zhang
Genes 2025, 16(5), 573; https://doi.org/10.3390/genes16050573 - 13 May 2025
Viewed by 610
Abstract
As the most aggressive primary brain tumor, glioblastoma (GBM) is considered incurable due to its molecular heterogeneity and therapy resistance. Identifying key regulatory factors in GBM is critical for developing effective therapeutic strategies. Based on the analysis of TCGA data, we confirmed a [...] Read more.
As the most aggressive primary brain tumor, glioblastoma (GBM) is considered incurable due to its molecular heterogeneity and therapy resistance. Identifying key regulatory factors in GBM is critical for developing effective therapeutic strategies. Based on the analysis of TCGA data, we confirmed a robust co-expression and correlation of OLIG1 and OLIG2 in human GBM. However, their roles in the astrocytic GBM subtype remain unclear. In this study, we first establish an astrocytic-featured GBM mouse model by introducing PiggyBac-driven hEGFRvIII plasmids and demonstrate that both OLIG1 and OLIG2 are highly expressed within this context. Next, using CRISPR/Cas9 technology to knockout Olig1/2, we found that astrocyte differentiation markers such as GFAP, SOX9, and HOPX were preserved, but tumor cell proliferation was significantly diminished. Mechanistically, CUT&Tag-seq revealed that OLIG1/2 directly binds to the promoter region of various cyclins (Cdk4, Ccne2, Ccnd3, and Ccnd1), where an enrichment of the active histone marker H3K4me3 was observed, indicating transcriptional activation of the genes. Notably, Olig1/2 knockout did not suppress tumor initiation or migration, suggesting that their primary role is to amplify proliferation rather than to drive tumorigenesis. This study defines Olig1 and Olig2 as master regulators of GBM proliferation through various cyclins, thereby offering a novel therapeutic target. Full article
(This article belongs to the Section Human Genomics and Genetic Diseases)
Show Figures

Figure 1

26 pages, 13470 KiB  
Article
Drug Combinations Targeting FAK and MEK Overcomes Tumor Heterogeneity in Glioblastoma
by Muhammad Furqan, Richard J. R. Elliott, Peter W. K. Nagle, John C. Dawson, Roza Masalmeh, Virginia Alvarez Garcia, Alison F. Munro, Camilla Drake, Gillian M. Morrison, Steven M. Pollard, Daniel Ebner, Valerie G. Brunton, Margaret C. Frame and Neil O. Carragher
Pharmaceutics 2025, 17(5), 549; https://doi.org/10.3390/pharmaceutics17050549 - 23 Apr 2025
Viewed by 1132
Abstract
Background/Objectives: Glioblastoma (GBM) is an aggressive brain tumor with limited treatment options and poor prognosis, largely owing to its heterogeneity and the involvement of multiple intracellular signaling pathways that contribute to drug resistance. While recent advancements in targeted drug combination therapies, such [...] Read more.
Background/Objectives: Glioblastoma (GBM) is an aggressive brain tumor with limited treatment options and poor prognosis, largely owing to its heterogeneity and the involvement of multiple intracellular signaling pathways that contribute to drug resistance. While recent advancements in targeted drug combination therapies, such as dabrafenib and trametinib, show promise for certain GBM subgroups, identifying effective drug combinations across the broader GBM population remains a challenge. Integrin-mediated signaling, particularly through Focal Adhesion Kinase (FAK), plays a pivotal role in GBM pathogenesis and invasion, making it a potential therapeutic target and component of future drug combination strategies. Methods: In this study, we utilized a chemogenomic screening approach to identify synergistic drug combinations that target FAK in glioblastoma. We initially employed a CRISPR-engineered GBM model to assess the effects of FAK depletion and subsequently discovered that combining FAK inhibitors such as VS4718 with MEK inhibitors, particularly trametinib, demonstrated synergistic effects. This potent combination was validated using various 2D and 3D assays, including cell viability/apoptosis assessment, synergistic analysis, cellular imaging, and target engagement assays. This combination also effectively inhibited spheroid growth and invasion across a diverse panel of patient-derived GBM stem cells. Molecular mechanisms underlying these effects include suppression of multiple kinase signaling pathways and enhanced apoptosis, elucidated using Reverse-Phase Protein Array (RPPA) profiling and Western blot validation. Result: In vivo, combination therapy significantly reduced the tumor volume in orthotopic transplantation models. Conclusions: These findings suggest that the combination of FAK and MEK inhibitors represents a promising therapeutic strategy to overcome the challenges of GBM treatment. Full article
(This article belongs to the Special Issue Combination Therapy Approaches for Cancer Treatment)
Show Figures

Figure 1

24 pages, 3012 KiB  
Article
Structural Activity Relationship Analysis of New Diphenyl PFI-3 Analogues Targeting for the Treatment of Glioblastoma
by Dong-Jin Hwang, Chuanhe Yang, Yinan Wang, Hannah Kelso, Satyanarayana Pochampally, Lawrence M. Pfeffer and Duane D. Miller
Pharmaceuticals 2025, 18(5), 608; https://doi.org/10.3390/ph18050608 - 23 Apr 2025
Cited by 1 | Viewed by 760
Abstract
Background/Objectives: Human glioblastoma (GBM) is the most aggressive brain cancer in adults and a highly treatment-refractory malignancy. The overall prognosis for the GBM is extremely poor, with a median survival of 12–14 months after initial diagnosis. Many GBM patients initially respond to [...] Read more.
Background/Objectives: Human glioblastoma (GBM) is the most aggressive brain cancer in adults and a highly treatment-refractory malignancy. The overall prognosis for the GBM is extremely poor, with a median survival of 12–14 months after initial diagnosis. Many GBM patients initially respond to the DNA alkylating agent temozolomide (TMZ), but patients often become therapy-resistant, and tumors recur. We previously reported that treatment with PFI-3, which is a small molecule inhibitor of the bromodomain of the BRG1 subunit of the SW1/SNF chromatin remodeling complex, enhanced the sensitivity of GBM cells to TMZ in vitro and in vivo GBM animal models. Our general objective was to perform an SAR study of new diphenyl PFI-3 analogs. Methods: New structural analogs of PFI-3 were developed, synthesized, and tested for their ability to enhance TMZ-induced GBM cell death by ELISA. Results: Following on the enhanced activity of compounds 2a and 2b, new diphenyl PFI-3 analogs with specific structural adjustments were made to better understand the structural requirements to optimize function. Additionally, several new structurally different candidates (e.g., 4a, 4b, and 5) showed much better efficacy in sensitizing GBM cells to TMZ-induced GBM cell death. Conclusions: Four series of PFI-3 analogs (2, 3, 4, and 5) were designed, synthesized, and tested for the ability to sensitize GBM cells to TMZ-induced cell death. Series 2 optimized the A-ring and R-isomer chirality. Series 3 used a 5-membered linker with weak activity. Series 4’s di-phenyl urea compounds showed better bromodomain inhibition. Series 5’s methoxyphenyl-B-ring analogs were exceptionally strong inhibitors. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Figure 1

20 pages, 1136 KiB  
Review
Kinase-Targeted Therapies for Glioblastoma
by Maria Salbini, Alessia Formato, Maria Patrizia Mongiardi, Andrea Levi and Maria Laura Falchetti
Int. J. Mol. Sci. 2025, 26(8), 3737; https://doi.org/10.3390/ijms26083737 - 15 Apr 2025
Viewed by 892
Abstract
Protein phosphorylation and dephosphorylation are key mechanisms that regulate cellular activities. The addition or removal of phosphate groups by specific enzymes, known as kinases and phosphatases, activates or inhibits many enzymes and receptors involved in various cell signaling pathways. Dysregulated activity of these [...] Read more.
Protein phosphorylation and dephosphorylation are key mechanisms that regulate cellular activities. The addition or removal of phosphate groups by specific enzymes, known as kinases and phosphatases, activates or inhibits many enzymes and receptors involved in various cell signaling pathways. Dysregulated activity of these enzymes is associated with various diseases, predominantly cancers. Synthetic and natural single- and multiple-kinase inhibitors are currently being used as targeted therapies for different tumors, including glioblastoma. Glioblastoma IDH-wild-type is the most aggressive brain tumor in adults, with a median overall survival of 15 months. The great majority of glioblastoma patients present mutations in receptor tyrosine kinase (RTK) signaling pathways responsible for tumor initiation and/or progression. Despite this, the multi-kinase inhibitor regorafenib has only recently been approved for glioblastoma patients in some countries. In this review, we analyze the history of kinase inhibitor drugs in glioblastoma therapy. Full article
(This article belongs to the Special Issue Latest Review Papers in Molecular Neurobiology 2025)
Show Figures

Graphical abstract

19 pages, 1118 KiB  
Review
Lactylation in Glioblastoma: A Novel Epigenetic Modifier Bridging Epigenetic Plasticity and Metabolic Reprogramming
by Qingya Qiu, Hui Deng, Ping Song, Yushu Liu and Mengxian Zhang
Int. J. Mol. Sci. 2025, 26(7), 3368; https://doi.org/10.3390/ijms26073368 - 4 Apr 2025
Cited by 3 | Viewed by 1612
Abstract
Glioblastoma, the most common and aggressive primary malignant brain tumor, is characterized by a high rate of recurrence, disability, and lethality. Therefore, there is a pressing need to develop more effective prognostic biomarkers and treatment approaches for glioblastoma. Lactylation, an emerging form of [...] Read more.
Glioblastoma, the most common and aggressive primary malignant brain tumor, is characterized by a high rate of recurrence, disability, and lethality. Therefore, there is a pressing need to develop more effective prognostic biomarkers and treatment approaches for glioblastoma. Lactylation, an emerging form of protein post-translational modification, has been closely associated with lactate, a metabolite of glycolysis. Since the initial identification of lactylation sites in core histones in 2019, accumulating evidence has shown the critical role that lactylation plays in glioblastoma development, assessment of poor clinical prognosis, and immunosuppression, which provides a fresh angle for investigating the connection between metabolic reprogramming and epigenetic plasticity in glioblastoma cells. The objective of this paper is to present an overview of the metabolic and epigenetic roles of lactylation in the expanding field of glioblastoma research and explore the practical value of developing novel treatment plans combining targeted therapy and immunotherapy. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

21 pages, 4743 KiB  
Article
Seco-Duocarmycin SA in Aggressive Glioblastoma Cell Lines
by Ann Morcos, Yeonkyu Jung, Ryan N. Fuller, Antonella Bertucci, Amy Nguyen, Quanqing Zhang, Tobias Emge, Kristopher E. Boyle, Nathan R. Wall and Marcelo Vazquez
Int. J. Mol. Sci. 2025, 26(6), 2766; https://doi.org/10.3390/ijms26062766 - 19 Mar 2025
Viewed by 675
Abstract
Glioblastoma multiforme (GBM) is among the most lethal primary brain tumors and is characterized by significant cellular heterogeneity and resistance to conventional therapies. This study investigates the efficacy of seco-duocarmycin SA (seco-DSA), a novel DNA alkylating agent. Initial investigations using a colony formation [...] Read more.
Glioblastoma multiforme (GBM) is among the most lethal primary brain tumors and is characterized by significant cellular heterogeneity and resistance to conventional therapies. This study investigates the efficacy of seco-duocarmycin SA (seco-DSA), a novel DNA alkylating agent. Initial investigations using a colony formation assay revealed that seco-DSA exhibits remarkable potential with IC50 values lower than its natural DSA counterpart. Cell viability assay indicated that LN18 cells showed a markedly greater sensitivity to DSA than T98G cells. Furthermore, seco-DSA achieved its full cytotoxic effect within 8 h of drug incubation in GBM cell lines. Although seco-DSA induced a concentration-dependent increase in apoptotic cell death, the extent of apoptosis did not fully account for the observed decrease in cell viability. Instead, seco-DSA treatment resulted in significant cell cycle arrest in S and G2/M phases. These findings suggest that seco-DSA’s cytotoxicity in GBM cells is primarily due to its ability to disrupt cell cycle progression, though the precise mechanisms of action remain to be fully established, and further research is needed. Proteomic analysis of treated cells also indicates dysregulation of proteins involved in senescence, apoptosis, and DNA repair, alluding to seco-DSA-induced arrest as a major mechanism of GBM disruption. Data are available via ProteomeXchange with the dataset identifier “PXD061023”. Our reports promote the future exploration of seco-DSA’s therapeutic potential, representing a critical step toward developing a more targeted and effective treatment for GBM. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

12 pages, 730 KiB  
Article
Dynamics Analysis of Nonlinear Differential Equation Systems Applied to Low-Grade Gliomas and Their Treatment
by Felipe J. Carmona-Moreno, Armando Gallegos, José J. Barba-Franco, Ernesto Urenda-Cázares, Enrique V. Jiménez-Guerrero and Jorge E. Macías-Díaz
Foundations 2025, 5(1), 6; https://doi.org/10.3390/foundations5010006 - 19 Feb 2025
Viewed by 1448
Abstract
Low-grade gliomas are a group of brain tumors that mostly affect people in early adulthood. A glioma is a tumor that originates in glial cells. They are classified into four levels according to their level of proliferation, with grades 1 and 2 called [...] Read more.
Low-grade gliomas are a group of brain tumors that mostly affect people in early adulthood. A glioma is a tumor that originates in glial cells. They are classified into four levels according to their level of proliferation, with grades 1 and 2 called low-grade gliomas. In this research, we conduct an analysis focused on a mathematical model that emulates the behavior of low-grade gliomas with chemotherapy treatment based on a system of nonlinear differential equations. An analysis of the model is carried out in the absence of treatment, resulting in a predictive solution for the behavior of glioma if it left untreated for some reason. In turn, a stability analysis is carried out on the system of equations to find the critical points for treating glioma. In addition, the numerical results of the model are obtained, presenting the state variables at each instant. Finally, some simulations are presented, varying the moments of treatment initiation and the applied doses of Temozolomide. Full article
(This article belongs to the Section Mathematical Sciences)
Show Figures

Figure 1

23 pages, 1783 KiB  
Article
Simultaneous Multi-Treatment Strategy for Brain Tumor Reduction via Nonlinear Control
by Muhammad Arsalan, Xiaojun Yu, Muhammad Tariq Sadiq and Ahmad Almogren
Brain Sci. 2025, 15(2), 207; https://doi.org/10.3390/brainsci15020207 - 17 Feb 2025
Viewed by 868
Abstract
Background: Recently proposed brain-tumor treatment strategies prioritize fast reduction of tumor cell population while often neglecting the radiation or chemotherapeutic drug dosage requirements to achieve it. Moreover, these techniques provide chemotherapy based treatment strategies, while ignoring the toxic side effects of the [...] Read more.
Background: Recently proposed brain-tumor treatment strategies prioritize fast reduction of tumor cell population while often neglecting the radiation or chemotherapeutic drug dosage requirements to achieve it. Moreover, these techniques provide chemotherapy based treatment strategies, while ignoring the toxic side effects of the drugs employed by it. Methods: This study updates the recently proposed brain-tumor system dynamics by incorporating radiotherapy along with chemotherapy to simultaneously initiate both therapies for a more comprehensive and effective response against tumor proliferation. Afterwards, based on the upgraded system dynamics, this study proposes a novel multi-input sigmoid-based smooth synergetic nonlinear controller with the aim to reduce the dosage requirements of both therapies while keeping the overall system response robust and efficient. The novelty of this study lies in the combination of radiotherapy and chemotherapy inputs in a way that prioritizes patients health and well-being, while integrating advanced synergetic control technique with a sigmoid function based smoothing agent. Results: The proposed method reduced baseline radiation and chemo drug dosages by 57% and 33% respectively while effectively suppressing tumor growth and proliferation. Similarly, the proposed controller reduced the time required for complete tumor mitigation by 60% while reducing the radiation and chemotherapeutic drug intensity by 93.8% and 21.3% respectively. Conclusions: This study offers significant improvement in tumor treatment methodologies by providing a safer, less riskier brain-tumor treatment strategy that has promising potential to improve survival rates against this menacing health condition so that the affected patients may lead a healthier and better quality of life. Full article
(This article belongs to the Special Issue Editorial Board Collection Series: Advances in Neuro-Oncology)
Show Figures

Figure 1

Back to TopTop