1
|
Gerster D, Muratoglu R, Giovannelli AC, Krahl P, Hansch A, Dieper A, Kaul D, Veltsista PD, Onken J, Misch M, Nadobny J, Zips D, Ghadjar P. Use of radiofrequency electromagnetic fields applied by capacitive hyperthermia for glioblastoma therapy. Int J Hyperthermia 2025; 42:2491518. [PMID: 40255192 DOI: 10.1080/02656736.2025.2491518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 03/31/2025] [Accepted: 04/06/2025] [Indexed: 04/22/2025] Open
Abstract
INTRODUCTION Radiofrequency electromagnetic fields applied by capacitive hyperthermia (cRF-HT) might be applicable to improve therapy for glioblastoma patients, but computer simulation data is scarce. We aimed to perform a numerical analysis of cRF-HT treatment in glioblastoma patients. METHODS The EHY-2030 cRF-HT system (Oncotherm, Budapest, Hungary) was studied using a round 20 cm diameter electrode. Realistic head models and quasi-electrostatic finite element simulations were created (Sim4Life v7.2, ZurichMedTech, Zürich, Switzerland). First, 109 spherical glioblastoma localizations were created within a healthy head model, and three different electrode setups were used to simulate the specific absorption rate (SAR). Then, in 20 real glioblastoma patients, the E-field and SAR in the gross tumor volume (GTV) and its boundary zone were simulated, and transient temperature simulations were performed. RESULTS The simulations conducted on 20 patients revealed that the SAR achieved in the GTV and its surrounding boundary zone is highly dependent on the localization of the tumor, with a mean SAR of 24.3 W/kg (ranging from 11.5 to 46.7 W/kg). The mean temperature within the GTV was higher in patients with a resection cavity (mean T50: 40.1 °C) instead of a macroscopic tumor (mean T50: 37.8 °C). The simulation outcome for the 109 artificial tumor localizations indicated enhanced effectiveness when the electrode is setup as close to the GTV as possible. CONCLUSION cRF-HT may induce mild hyperthermia in a subgroup of glioblastoma patients with resection cavities. In macroscopic tumors, temperatures remain below the hyperthermia threshold. Further research is required to assess the clinical benefit of this therapy.
Collapse
Affiliation(s)
- Dominik Gerster
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Rami Muratoglu
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Paul Krahl
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Alexander Hansch
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Anna Dieper
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - David Kaul
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Paraskevi D Veltsista
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Onken
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Martin Misch
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jacek Nadobny
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Daniel Zips
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Pirus Ghadjar
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
2
|
Morshedi B, Esfandyari-Manesh M, Atyabi F, Ghahremani MH, Dinarvand R. Local delivery of ibrutinib by folate receptor-mediated targeting PLGA-PEG nanoparticles to glioblastoma multiform: in vitro and in vivo studies. J Drug Target 2025; 33:1026-1041. [PMID: 39960788 DOI: 10.1080/1061186x.2025.2468749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/19/2025] [Accepted: 02/09/2025] [Indexed: 02/25/2025]
Abstract
Glioblastoma multiforme (GBM) is a widespread and life-threatening kind of brain cancer, which has a high mortality rate. Ibrutinib, a Bruton's tyrosine kinase (BTK) inhibitor, irreversibly adheres to a conserved cysteine residue of two enzymes BTK and BMX, inhibiting their kinase activities, which leads to suppression of the growth of glioma cells. This study synthesised PLGA-PEG-folate (PPF) polymer and subsequently encapsulated ibrutinib within PPF nanoparticles (IBT-PPF-NPs). H NMR spectra confirmed the synthesis of PPF polymer. The efficiency of IBT-PPF-NPs was 97 ± 2.26% with 8.8 ± 0.2% drug loading. The particle size was 208 ± 4.8 nm. The IC50 value of free ibrutinib, IB-PPF-NPs and ibrutinib encapsulated in PLGA NPs (IB-P-NPs) was 10.2, 7.6 and 10.13 µM in C6 cell lines, whereas in U-87 MG cells was 24.4, 16 and 25.2 µM, respectively. The cellular uptake of FITC-PPF-NPs increased from 47.6% to 90.3% in C6 cells and from 55% to 97.3% in U-87 MG cells compared to FITC-P-NPs. The in vivo results indicate a significant reduction in tumour size in treatment groups in comparison to control groups, while the group that received the intratumoural injection of IB-PPF-NPs exhibited a greater reduction. The folate-targeting agent enhances the nanoparticles' effectiveness by promoting their uptake through the endocytosis pathway.
Collapse
Affiliation(s)
- Bahar Morshedi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Esfandyari-Manesh
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Atyabi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Ghahremani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Rassoul Dinarvand
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Dasgupta P, Puduvalli VK. Diversity of metabolic features and relevance to clinical subtypes of gliomas. Semin Cancer Biol 2025; 112:126-134. [PMID: 40194749 DOI: 10.1016/j.semcancer.2025.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 02/13/2025] [Accepted: 03/31/2025] [Indexed: 04/09/2025]
Abstract
Gliomas carry a dismal prognosis and have proven difficult to treat. Current treatments and efforts to target individual signaling pathways have failed. This is thought to be due to genetic and epigenetic heterogeneity and resistance. Therefore, interest has grown in developing a deeper understanding of the metabolic alterations that represent drivers and dependencies in gliomas. Therapies that target glioma-specific metabolic dependencies overcome the challenges of disease heterogeneity. Here, we present the diverse metabolic features of each current clinical subtype of glioma. We believe that this approach will enable the development of novel strategies to specifically target the various clinical and molecular subtypes of glioma using these metabolic features.
Collapse
Affiliation(s)
- Pushan Dasgupta
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA.
| | - Vinay K Puduvalli
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA.
| |
Collapse
|
4
|
Ma C, Wang J, Li Q, Wu Y, Yu Z, Chao Y, Liu Z, Chen G. Injectable oxidized high-amylose starch hydrogel scaffold for macrophage-mediated glioblastoma therapy. Biomaterials 2025; 318:123128. [PMID: 39884130 DOI: 10.1016/j.biomaterials.2025.123128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/19/2025] [Accepted: 01/23/2025] [Indexed: 02/01/2025]
Abstract
Glioblastoma, characterized by rapid proliferation and invasiveness, is largely resistant to current treatment modalities. A major obstacle is the blood-brain barrier (BBB), which restricts the delivery of therapeutic agents as well as the infiltration of effective immune cells into glioblastoma. In this study, we developed an injectable oxidized high-amylose starch hydrogel (OHASM) to serve as a biomaterial scaffold for the delivery of macrophages and macrophage-polarizing drugs, aiming to bypass the BBB and enhance glioblastoma treatment. The in vitro and in vivo experiments confirmed the efficacy of the hydrogel in loading and delivering macrophages and polarizing drugs against glioblastoma. Additionally, the hydrogel's interconnected porous structure was conducive to cellular growth and activity, and its slow release of therapeutics contributed to the extended survival of treated mice in a mouse GL261 glioblastoma tumor model. The immunological mechanisms underlying the therapeutic efficacy were further elucidated, revealing the potential of the hydrogel system to modulate macrophage polarization and induce apoptosis in tumor cells via the poly ADP-ribose polymerase (PARP) pathway. The study underscores the potential of the hydrogel-based macrophage delivery strategy as an effective and safe treatment for glioblastoma, offering a promising avenue for clinical management of this aggressive brain cancer.
Collapse
Affiliation(s)
- Cheng Ma
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006 China
| | - Jiahe Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006 China
| | - Qiaofeng Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123 China
| | - Yuzhe Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123 China
| | - Zhengquan Yu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006 China
| | - Yu Chao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123 China.
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123 China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006 China.
| |
Collapse
|
5
|
Maleszewska M, Roura AJ, Dabrowski MJ, Draminski M, Wojtas B. Decoding glioblastoma's diversity: Are neurons part of the game? Cancer Lett 2025; 620:217666. [PMID: 40147584 DOI: 10.1016/j.canlet.2025.217666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
Glioblastoma multiforme (GBM, WHO Grade 4) is a highly aggressive primary brain tumor with limited treatment options and a poor prognosis. A key challenge in GBM therapy lies in its pronounced heterogeneity, both within individual tumors (intratumoral) and between patients (intertumoral). Historically, neurons have been underexplored in GBM research; however, recent studies reveal that GBM development is closely linked to neural and glial progenitors, often mimicking neurodevelopmental processes in a dysregulated manner. Beyond damaging neuronal tissue, GBM actively engages with neurons to promote pro-tumorigenic signaling, including neuronal hyperexcitability and seizures. Single-cell RNA sequencing (scRNA-seq) has revolutionized our understanding of the tumor microenvironment (TME), uncovering the critical roles of immune cells, endothelial cells, and astrocytes in tumor progression. However, technical limitations of scRNA-seq hinder its ability to capture the transcriptomes of neurons, necessitating the use of single-nucleus RNA sequencing (snRNA-seq) to study these interactions at single-cell resolution. This work collects the emerging insights of glioblastoma-neuron interactions, focusing on how GBM exploits neurodevelopmental pathways and reshapes neuronal networks. Moreover, we perform bioinformatic analysis of publicly available snRNA-seq datasets to propose putative cell-cell interactions driving glioma-neuronal dynamics. This study delineates key signaling pathways and underscores the need for further investigation to evaluate their potential as therapeutic targets.
Collapse
Affiliation(s)
- Marta Maleszewska
- Department of Animal Physiology, Institute of Experimental Zoology, Faculty of Biology, University of Warsaw, 1 Miecznikowa Str, 02-096, Warsaw, Poland.
| | - Adrià-Jaume Roura
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
| | - Michal J Dabrowski
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| | - Michal Draminski
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| | - Bartosz Wojtas
- Laboratory of Sequencing, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
6
|
Cheng C, Cui L, Cui X, Zhan Q, Ju J, Hong B, Huang Y, Ding Y, Xu H, Qiu T, Kang C, Liu X, Wang Q, Zeng L. ADAM12 promotes temozolomide resistance in glioblastoma by activating the TNF-α - NF-κB pathway. Cancer Lett 2025; 620:217684. [PMID: 40180114 DOI: 10.1016/j.canlet.2025.217684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/05/2025]
Abstract
Development of temozolomide (TMZ) resistance is a critical factor contributing to a poor prognosis in glioma patients. TMZ resistance is also closely associated with the phosphorylation level of NF-κB, yet targeted inhibition of NF-κB activity in glioma can be leveraged to overcome TMZ resistance. ADAM12, a protein significantly overexpressed in glioma cells, is implicated in the pathogenesis and progression of glioma, yet its role in the development of TMZ resistance is completely understood. We found that knockdown of ADAM12 was shown to arrest the glioma cell cycle, enhance apoptosis, inhibit DNA damage repair mechanisms, and sensitize glioma cells to TMZ. Targeting ADAM12 in vivo was found to increase the sensitivity of glioma cells to TMZ. Survival analysis indicated that ADAM12 serves as a prognostic marker for TMZ treatment. Using ELISA and protein interaction predictions via docking simulation, we identified the TNF-α shedding function of ADAM12 as a critical regulator of glioma progression. Furthermore, in glioma cell lines with unmethylated MGMT, the knockdown of ADAM12 enhanced sensitivity to TMZ by inhibiting the TNF-α/NF-κB pathway and reducing MGMT expression. In all, these results demonstrated that ADAM12 aids in shedding of membrane-bound TNF-a to drive TMZ resistance in glioma.
Collapse
Affiliation(s)
- Chunchao Cheng
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Longtao Cui
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xiaoteng Cui
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Qi Zhan
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jiasheng Ju
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Biao Hong
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yanping Huang
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yaqing Ding
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Hanyi Xu
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Tian Qiu
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Chunsheng Kang
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xiaomin Liu
- Neuro-Oncology Center, Tianjin Huanhu Hospital, Tianjin, 300350, China.
| | - Qixue Wang
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital and Key Laboratory of Neurotrauma, Variation, and Regeneration, Ministry of Education and Tianjin Municipal Government, Tianjin, 300052, China; Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Liang Zeng
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
7
|
Huang J, Yu H, Wu J, Yin B, Hou L, Qiang B, Shu P, Peng X. ZBED6 inhibits the migration of glioblastoma cells by regulating the Wnt/β-catenin signaling pathway. Biochem Biophys Res Commun 2025; 766:151830. [PMID: 40306163 DOI: 10.1016/j.bbrc.2025.151830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2025] [Accepted: 04/15/2025] [Indexed: 05/02/2025]
Abstract
ZBED6, a member of the zinc-finger protein superfamily, has incompletely defined biological functions in cancer. Previous studies suggest that ZBED6 may influence colorectal cancer growth and cell cycle regulation. This study aims to investigate the biological role and regulatory mechanisms of ZBED6 in glioblastoma. Bioinformatics analysis revealed abnormal activation of the Wnt/β-catenin signaling pathway in glioblastoma and a negative correlation between ZBED6 expression and Wnt/β-catenin signaling. Survival analysis further indicated that low ZBED6 expression is associated with poor prognosis in GBM patients. Through in vitro experiments, we demonstrated that ZBED6, as a transcription factor, regulates its downstream target TCF7L2, thereby modulating the Wnt/β-catenin signaling pathway. Additionally, we found that ZBED6 acts as a critical negative regulator of glioblastoma migration. Loss of ZBED6 expression may drive malignant progression by activating mesenchymal transition signaling, alleviating cell cycle arrest, and promoting oncogenic pathway activation. Collectively, these findings suggest that reduced ZBED6 expression in glioblastoma leads to aberrant TCF7L2 expression and dysregulated Wnt/β-catenin signaling activation, which is closely linked to poor clinical outcomes in GBM.
Collapse
Affiliation(s)
- Junxiao Huang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry & Molecular Biology, Medical Primate Research Center, Neuroscience Center, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Haoyang Yu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry & Molecular Biology, Medical Primate Research Center, Neuroscience Center, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Jiarui Wu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry & Molecular Biology, Medical Primate Research Center, Neuroscience Center, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Bin Yin
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry & Molecular Biology, Medical Primate Research Center, Neuroscience Center, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Lin Hou
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry & Molecular Biology, Medical Primate Research Center, Neuroscience Center, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Boqin Qiang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry & Molecular Biology, Medical Primate Research Center, Neuroscience Center, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China
| | - Pengcheng Shu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry & Molecular Biology, Medical Primate Research Center, Neuroscience Center, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China.
| | - Xiaozhong Peng
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry & Molecular Biology, Medical Primate Research Center, Neuroscience Center, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China; State Key Laboratory of Respiratory Health and Multimorbidity, Beijing, 100005, China; Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
8
|
Kim YB, Lee SH, Kasala D, Zhao Y, Jiao A, Hong J, Kim JS, Yoon AR, Yun CO. Potent therapeutic efficacy of intranasally deliverable paclitaxel modified with pH-sensitive and PEGylated polymeric micelle against glioblastoma. J Control Release 2025; 382:113711. [PMID: 40204132 DOI: 10.1016/j.jconrel.2025.113711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/10/2025] [Accepted: 04/06/2025] [Indexed: 04/11/2025]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive and common type of brain tumor. Conventional therapies for GBM, such as surgery or radiotherapy, have shown inadequate therapeutic effect. Similarly, a large fraction of chemotherapeutics are ineffective against GBM due to the blood-brain barrier (BBB) preventing effective delivery of these drugs to the brain. To overcome these obstacles, an intranasally administrable and multifunctional drug-loaded polymeric micelle composed of a pH-sensitive PPCBA-PEI-Arg (PPA) polymer conjugated with PEGylated paclitaxel (PEG-PTX; PPP) was synthesized to treat GBM. PPP was more soluble in an aqueous solution than parental PTX and was more effectively internalized into the GBM cells. Further, PPP elicited a more potent cancer cell killing effect than PTX under physiological pH condition, which was further augmented under the mildly acidic condition that emulated the tumor microenvironment. Intranasal administration of PPP into orthotopic GBM tumor xenograft-bearing mice led to more efficient delivery of the drug to the brain tissues compared to parental PTX delivered via intranasal or intravenous route, thus resulting in superior inhibition of GBM growth. Collectively, these findings demonstrated that intranasal delivery of PTX via pH-sensitive and PEGylated polymeric micelles can be an effective approach for the treatment of aggressive GBM.
Collapse
Affiliation(s)
- Young-Beom Kim
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea
| | - Soo-Hwan Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea
| | - Dayananda Kasala
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea
| | - Yuebin Zhao
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea
| | - Ao Jiao
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea
| | - JinWoo Hong
- GeneMedicine Co., Ltd., Seoul, Republic of Korea
| | - Jin Su Kim
- Radiological and Medico-Oncological Sciences, University of Science and Technology (UST), Seoul, Republic of Korea
| | - A-Rum Yoon
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea; Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, Republic of Korea; Institute of Nano Science and Technology (INST), Hanyang University, Seoul, Republic of Korea
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea; Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, Republic of Korea; Institute of Nano Science and Technology (INST), Hanyang University, Seoul, Republic of Korea; GeneMedicine Co., Ltd., Seoul, Republic of Korea.
| |
Collapse
|
9
|
Man S, Li J, Li Y, Yan F, Wang Z, Huang J, Xia Y, Khan AJ, Wang L, Jia S, Wang J, Liu X, Zhang Y, Rahman FU, Li X. Elucidating the dual mechanistic action and synergism of platinum complexes bearing valproic acid as leaving ligand on NF-κB and inflammatory pathways in glioma. Eur J Med Chem 2025; 290:117522. [PMID: 40153933 DOI: 10.1016/j.ejmech.2025.117522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/27/2025] [Accepted: 03/16/2025] [Indexed: 04/01/2025]
Abstract
The valproic acid (VPA), an anti-epileptic drug, has demonstrated anticancer properties alone or in combination regimens in glioma. It has been shown synergistic activity with cisplatin in resistant cancer cells. In the current study, we synthesized Pt(II) complexes bearing VPA as ancillary/leaving ligand. All these complexes were obtained in good yields through simple reproducible synthetic procedures and characterized by multiple analytical techniques in both solution and solid state. In situ release of ancillary ligand (VPA) by these complexes was studied by 1H NMR in solution state that was catalysed by water in time dependent manner. The tumor preferential selective VPA-Pt actively controlling NF-kB signaling, culminating in the attenuation of IL-6 expression and the concomitant activation of p53 and caspase-3 pathways in gliomas. VPA-Pt exhibits potent cytotoxicity in human and mice glioma cancer cell lines, inducing apoptosis as evidenced by inhibition of cell proliferation and migration, disruption of mitochondrial membrane potential, and suppression of colony formation. An inhibitory effect of VPA-Pt4 on glioma was clearly evidenced through in vivo live bioluminescence imaging, histopathological examination, immunofluorescence evaluation, and protein expression analysis demonstrated that VPA-Pt4 significantly triggered apoptosis, with elevated levels of P53, caspase-3, cleaved caspase-3, along with a reduction in IL-6. Our discovery reveals a novel and efficient approach to glioma therapy.
Collapse
Affiliation(s)
- Shad Man
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot, 010021, People's Republic of China; Inner Mongolia University Research Center for Glycochemistry of Characteristic Medicinal Resources, Department of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot, 010021, People's Republic of China
| | - Jiaqi Li
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot, 010021, People's Republic of China
| | - Yimiao Li
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot, 010021, People's Republic of China
| | - Fufu Yan
- Inner Mongolia University Research Center for Glycochemistry of Characteristic Medicinal Resources, Department of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot, 010021, People's Republic of China
| | - Zerui Wang
- Inner Mongolia University Research Center for Glycochemistry of Characteristic Medicinal Resources, Department of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot, 010021, People's Republic of China
| | - Jinxia Huang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot, 010021, People's Republic of China
| | - Yan Xia
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot, 010021, People's Republic of China
| | - Abdul Jamil Khan
- Biomedical Nanocenter, School of Life Science, Inner Mongolia Agricultural University, Hohhot, 010018, People's Republic of China
| | - Liping Wang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot, 010021, People's Republic of China
| | - Shuang Jia
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot, 010021, People's Republic of China
| | - Jie Wang
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot, 010021, People's Republic of China
| | - Xing Liu
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot, 010021, People's Republic of China
| | - Yongmin Zhang
- Inner Mongolia University Research Center for Glycochemistry of Characteristic Medicinal Resources, Department of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot, 010021, People's Republic of China; Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, UMR 8232, 4 Place Jussieu, 75005, Paris, France
| | - Faiz-Ur Rahman
- Inner Mongolia University Research Center for Glycochemistry of Characteristic Medicinal Resources, Department of Chemistry and Chemical Engineering, Inner Mongolia University, Hohhot, 010021, People's Republic of China.
| | - Xinyu Li
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, Inner Mongolia University, Hohhot, 010021, People's Republic of China.
| |
Collapse
|
10
|
Zhang S, McMillan N, McGuinness M, Trudeau S, Ho KWG, Uhlmann EJ. Peripheral biomarkers predict survival in patients with glioblastoma treated with temozolomide. Mol Clin Oncol 2025; 22:56. [PMID: 40322547 PMCID: PMC12046623 DOI: 10.3892/mco.2025.2851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/04/2024] [Indexed: 05/08/2025] Open
Abstract
Grade 4 gliomas, including glioblastoma, isocitrate dehydrogenase (IDH)-wild-type [GBM (IDH wt)], and astrocytoma, IDH-mutant, grade 4, are among the most aggressive primary brain tumors. The standard of care for GBM (IDH wt) and astrocytoma, IDH-mutant, grade 4 is maximum safe resection and radiation plus oral temozolomide (TMZ) followed by six cycles of TMZ. The study objective was to identify peripheral biomarkers that predict favorable outcomes and stratify patients likely to respond to treatment. Adults with biopsy-confirmed glioblastoma (based upon the 2016 World Health Organization classification) treated at Beth Israel Deaconess Medical Center (Boston, USA) between January 2018 and November 2021 were identified. Data on laboratory values (white blood cells, absolute neutrophil count, absolute lymphocyte count, red blood cells, hemoglobin and platelet count), molecular markers, including IDH1 R132H and methylguanine-DNA methyltransferase promoter methylation, and progression-free survival (PFS) and overall survival (OS) were collected retrospectively. Data were combined with those from two prior studies, resulting in a total of 263 patients. Leukopenia development during TMZ treatment was associated with increased PFS (P=0.008) and OS (P=0.03). Lymphopenia development during TMZ treatment was associated with increased PFS (P=0.05) and OS (P=0.007). Grade 3 thrombocytopenia during TMZ treatment was associated with decreased PFS (P=0.01) and OS (P=0.02). Patients who developed leukopenia alone during treatment had an increased OS compared with those with only lymphopenia development and those with both lymphopenia and leukopenia development (P=0.007). Lower baseline lymphocyte counts (<0.7 K/µl) prior to treatment was associated with improved OS (P=0.007), while increased baseline neutrophil counts (≥10.0 K/µl) prior to initiation of treatment were associated with worse OS (P=0.002). In conclusion, TMZ exposure may result in a leukocyte predominant bone marrow effect vs. a platelet predominant bone marrow effect. Clinically, leukopenia could indicate adequate TMZ dosing, with thrombocytopenia serving as a limiting factor in the ability to continue TMZ. Baseline counts may offer insights into which patients will benefit the most from treatment.
Collapse
Affiliation(s)
- Shuodan Zhang
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Nadia McMillan
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Matthew McGuinness
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Stephen Trudeau
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ka-Wai Grace Ho
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Erik J. Uhlmann
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
11
|
Brenner DA, Dadario NB, Zaman A, Valdivia DJ, Pandya M, Yeung J, Sughrue M, Teo C. Surgical outcomes in high-grade adult type diffuse gliomas (ATDG) with a previous diagnosis of anaplastic astrocytoma without adjuvant therapy. Clin Neurol Neurosurg 2025; 253:108879. [PMID: 40253838 DOI: 10.1016/j.clineuro.2025.108879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 03/28/2025] [Accepted: 03/29/2025] [Indexed: 04/22/2025]
Abstract
INTRODUCTION Gliomas pose a significant treatment challenge due to their varied genetic makeup and clinical presentations. This study examines a unique cohort of high-grade adult type diffuse gliomas (ATDG) previously diagnosed as anaplastic astrocytoma prior to the WHO 2021 tumor classification changes. This cohort chose to undergo only surgical resection without adjuvant therapies. We provide a rare dataset of patients allowing for new insight into the natural progression of this disease with surgical treatment alone. METHODS A retrospective review was conducted of patients who were operated on by a single surgeon from the years 2002-2022 and who were diagnosed as having a Grade III Anaplastic Astrocytoma before the WHO 2021 guidelines were published. Correcting for the criteria in the 2021 Guidelines resulted in a mixture of adult-type diffuse malignant gliomas (ATDG), including IDH-Mutant astrocytomas (Grade 3 and 4) and IDH-WT Glioblastoma. All patients included underwent surgical resection alone after declining any adjuvant therapy for various reasons. RESULTS A total of 20 patients met the inclusion criteria with an average age of 38 years. Among them, 15 had IDH-mutant (IDH-mt) Grade 3 astrocytomas (75 %), 1 had an IDH-mt Grade 4 astrocytoma (5 %), and 4 had IDH-wildtype (IDH-WT) glioblastomas (20 %). The 5-year survival rate for the entire cohort was 74.0 %. Grade 3 astrocytomas had a 5-year survival of 86.7 %, while Grade 4 astrocytomas and IDH-WT GBM patients exhibited a 5-year survival rate of 40 %. 5-year progression-free survival (PFS) rates were derived from the surgery date up until the recurrence or censorship. The collective cohort had a PFS rate of 34.3 %. Grade 3 astrocytomas achieved a 5-year PFS of 32.0 %, whereas Grade 4 astrocytomas and IDH-WT GBM reached a PFS of 40.0 %. CONCLUSION In our cohort study, we demonstrate that patients with ATDG can potentially achieve relative long-term survival through surgical resection alone. This unique cohort highlights the natural progression of this disease with surgery alone and provides the foundation for future more rigorous studies to evaluate the additive benefit of different adjuvant therapies. With evolving tumor classifications and variable responses to standard therapeutics, it becomes imperative to revisit and understand the additive benefits of different chemotherapeutic protocols in addition to surgical resection.
Collapse
Affiliation(s)
- Daniel A Brenner
- Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Nicholas B Dadario
- Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Ashraf Zaman
- Garvan Institute of Medical Research, Sydney, Australia; Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Daniel J Valdivia
- Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | | | - Jacky Yeung
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | | | | |
Collapse
|
12
|
Xiong Y, He C, Qi J, Xiong M, Liu S, Zhao J, Li Y, Liu G, Deng W. Black phosphorus nanosheets activate tumor immunity of glioblastoma by modulating the expression of the immunosuppressive molecule PD-L1. Biomaterials 2025; 317:123062. [PMID: 39736218 DOI: 10.1016/j.biomaterials.2024.123062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/22/2024] [Accepted: 12/26/2024] [Indexed: 01/01/2025]
Abstract
The tumor microenvironment in glioblastoma (GBM) is characterized by a pronounced immunosuppressive state, which significantly hampers tumor treatment and contributes to treatment resistance. While our previous research established that black phosphorus nanosheets (BPNS) inhibited glioblastoma cell migration and invasion, the impact of BPNS on the anti-tumor-associated immune mechanism remains unexplored. This study firstly investigated whether BPNS could modulate the tumor microenvironment through immunotherapy and elucidated the underlying mechanisms. We used a subcutaneous mouse model of GBM, which evaded immune surveillance to evaluate BPNS effects on immune cells within the tumor microenvironment. Our results demonstrated that BPNS significantly enhanced the tumor-suppressive microenvironment, reactivating immune cells' cytotoxicity against tumor cells. Moreover, further analysis revealed that BPNS counteracted the immunosuppressive state by reducing the expression of the immunosuppressive molecule PD-L1 in tumor cells, leading to an anti-tumor effect. Mechanistically, BPNS reduced PD-L1 expression through two main pathways: by inducing autophagy via binding to the HSP90 protein, leading to PD-L1 degradation through the autophagy pathway, and by inhibiting the PI3K-AKT signaling pathway, which reduced PD-L1 mRNA levels. This study expands the understanding of BPNS biological activity and suggests new strategies for utilizing BPNS as an adjuvant in immunotherapy.
Collapse
Affiliation(s)
- Yue Xiong
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; The Eighth Affiliated Hospital, Sun Yat-sen University, 3025 Shennan Middle Road, Shenzhen, 518000, China
| | - Chao He
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Junyang Qi
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Meimei Xiong
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Shuna Liu
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Jingxin Zhao
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yuzhen Li
- The Eighth Affiliated Hospital, Sun Yat-sen University, 3025 Shennan Middle Road, Shenzhen, 518000, China
| | - Gan Liu
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China.
| | - Wenbin Deng
- School of Pharmaceutical Sciences, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
13
|
Li H, Liu Y, Zhang H, Wang X. DEP domain‑containing 1 is a prognostic biomarker associated with the cell cycle in gliomas. Oncol Lett 2025; 29:276. [PMID: 40247989 PMCID: PMC12005072 DOI: 10.3892/ol.2025.15022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 03/11/2025] [Indexed: 04/19/2025] Open
Abstract
Glioma is the most common primary tumor in the intracranial region, accounting for more than one-half of all central nervous system tumors. Abnormal expression of key cancer genes often promotes the occurrence and development of tumors. DEP domain containing 1 (DEPDC1) is a gene that encodes a protein containing a DEP domain, which serves an important role in numerous biological processes. In the present study, the relationship between the expression level of DEPDC1 and the clinical features of glioma was explored in datasets from the China Glioma Genome Atlas Project. Kaplan-Meier survival analysis was performed to evaluate the value of DEPDC1 expression in the prognosis of patients with glioma. T-test and univariate Cox analysis were used to identify differential genes, and Gene Ontology and gene set enrichment analysis (GSEA) were used to explore the function and related mechanisms of DEPDC1 in glioma. Univariate cox and multivariate cox analyses were used to screen variables, and a nomogram model was used to construct a prediction model. In glioma U87 and LN229 cell lines, the expression of DEPDC1 was decreased using shRNA to assess the effects of DEPDC1 on the proliferation, migration and invasion of glioma cells. The findings revealed that there was a positive association between the expression level of DEPDC1 and the poor clinical features of glioma, and patients with high expression of DEPDC1 had a significantly shorter overall survival time. GSEA demonstrated that the differential genes in the DEPDC1 high expression group were mainly enriched in 'cell cycle' and 'mitotic cell cycle'. Cell experiments showed that silencing DEPDC1 in U87 and LN229 cells significantly attenuated cell proliferation, migration and invasion. To conclude, the present study demonstrates that DEPDC1 is an independent prognostic indicator for patients with glioma and is associated with a poor prognosis. The expression of DEPDC1 is closely associated with the cell cycle of glioma and provides individualized treatment options for tumors.
Collapse
Affiliation(s)
- Haima Li
- Department of Neurosurgery, Shaanxi Nuclear Industry 215 Hospital, Xianyang, Shaanxi 712000, P.R. China
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Yezu Liu
- Department of Neurosurgery, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Hanwen Zhang
- Department of Neurosurgery, Peking University People's Hospital, Beijing 100044, P.R. China
| | - Xuelian Wang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| |
Collapse
|
14
|
Lee SH, Hofstede RP, Noriega de la Colina A, Gunton JH, Bernstock JD, Traverso G. Implantable systems for neurological chronotherapy. Adv Drug Deliv Rev 2025; 221:115574. [PMID: 40187646 DOI: 10.1016/j.addr.2025.115574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/26/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025]
Abstract
Implantable systems for neurological chronotherapy are poised to revolutionize the treatment of central nervous system diseases and disorders. These devices enable precise, time-controlled drug delivery aligned with the body's circadian rhythms, optimizing therapeutic outcomes. By bypassing the blood-brain barrier, they achieve high local drug concentrations while minimizing systemic side effects, offering significant advantages for conditions where traditional therapies often fall short. Platforms like SynchroMed II and CraniUS showcase this innovation, providing programmable delivery for conditions such as epilepsy and glioblastoma, with customizable profiles ranging from continuous infusion to timed bolus administration. Preclinical and clinical studies underscore the efficacy of aligning drug delivery with circadian rhythms, enhancing outcomes in chrono-chemotherapy and anti-epileptic treatments. Despite their promise, challenges remain, including the invasiveness of implantation within the brain, device longevity, synchronization complexities, and cost(s). Accordingly, this review explores the current state of implantable neurological systems that may be leveraged for chronotherapy, their applications, limitations, and potential to transform neurological disease/disorder management.
Collapse
Affiliation(s)
- Seung Ho Lee
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Roemer Pott Hofstede
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - John H Gunton
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Joshua D Bernstock
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Giovanni Traverso
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
15
|
Bhangale P, Kashikar S, Parihar PH, Patil R, Gupta V, Shinde D, Nimodia D. A rare case of gliosarcoma: Comprehensive radiological, histopathological, and clinical insights into diagnosis and management. Radiol Case Rep 2025; 20:2676-2681. [PMID: 40151286 PMCID: PMC11937613 DOI: 10.1016/j.radcr.2025.02.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 03/29/2025] Open
Abstract
Gliosarcoma is a rare and aggressive variant of glioblastoma, characterized by a biphasic histological pattern consisting of both glial and mesenchymal components. This case report describes the clinical presentation, radiological findings, surgical management, and histopathological analysis of gliosarcoma in a 30-year-old female. The patient presented with a 10-day history of right-sided headache and recurrent vomiting. Neurological examination was unremarkable, and vital signs were stable. Magnetic resonance imaging (MRI) revealed a heterogeneously enhancing mass lesion involving the right parietal region and the splenium of the corpus callosum, crossing the midline and causing significant ventricular effacement. Imaging features included heterogeneously hypointense signals on T1-weighted imaging, hyperintense signals on T2/FLAIR, areas of blooming on susceptibility-weighted imaging, and restricted diffusion on diffusion-weighted imaging, suggestive of a high-grade glial tumor. The patient underwent surgical resection, and histopathological examination confirmed gliosarcoma. The tumor exhibited a biphasic pattern comprising glial and sarcomatous elements. This case emphasizes the diagnostic challenges associated with gliosarcoma, where radiological features often mimic glioblastoma, necessitating histopathological confirmation. Gliosarcoma's aggressive nature poses significant therapeutic challenges, with treatment strategies involving surgical resection followed by adjuvant radiotherapy and chemotherapy. This report highlights the importance of integrating clinical, radiological, and histopathological findings to achieve an accurate diagnosis and optimize treatment outcomes. It underscores the need for early recognition and a multidisciplinary approach to managing rare central nervous system tumors like gliosarcoma. Further research into advanced therapeutic strategies is warranted to improve the prognosis for such patients.
Collapse
Affiliation(s)
- Paritosh Bhangale
- Department of Radiodiagnosis, Datta Meghe Institute of Medical Sciences, Wardha, Maharashtra, India, 442001
| | - Shivali Kashikar
- Department of Radiodiagnosis, Datta Meghe Institute of Medical Sciences, Wardha, Maharashtra, India, 442001
| | | | - Ravishankar Patil
- Department of Radiodiagnosis, Datta Meghe Institute of Medical Sciences, Wardha, Maharashtra, India, 442001
| | - Viraj Gupta
- Department of Radiodiagnosis, Datta Meghe Institute of Medical Sciences, Wardha, Maharashtra, India, 442001
| | - Dhananjay Shinde
- Department of Radiodiagnosis, Datta Meghe Institute of Medical Sciences, Wardha, Maharashtra, India, 442001
| | - Devyansh Nimodia
- Department of Radiodiagnosis, Datta Meghe Institute of Medical Sciences, Wardha, Maharashtra, India, 442001
| |
Collapse
|
16
|
Hama Y, Sasaki T, Fukai J, Nakao N. Bevacizumab‑associated intracerebral hemorrhage in patients with malignant glioma. Mol Clin Oncol 2025; 22:57. [PMID: 40322545 PMCID: PMC12046618 DOI: 10.3892/mco.2025.2852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/11/2025] [Indexed: 05/08/2025] Open
Abstract
Intracerebral hemorrhage (ICH) is a serious complication of the use of bevacizumab in patients with malignant glioma; however, the risk factors are unclear. Therefore, the present study retrospectively analyzed a cohort of patients treated with bevacizumab for malignant glioma to investigate the characteristics of those in the cohort who had ICH. Between January 2015 and December 2022, 64 patients with malignant glioma were treated with bevacizumab. Clinical and molecular biological information, treatment details, and information regarding the presence of ICH after bevacizumab administration were extracted from the hospital database. ICH was found to have occurred in seven patients (10.9%) after bevacizumab administration. The mean (standard deviation) age of these seven patients was 64(11) years, and six of them (85.7%) underwent needle biopsy. Two patients (28.6%) had grade ≥3 hemorrhage. The median number of administrations of bevacizumab before the onset of ICH was seven (range: 1-32), and the duration from first administration to ICH was 4 months (range: 1-22). Furthermore, ICH was associated with a comparatively short overall survival time (log-rank, P=0.008). Tumor invasion into the corpus callosum on contrast-enhanced magnetic resonance imaging before bevacizumab administration was associated with ICH according to univariate analysis (P=0.01) and multivariate analysis (P=0.02). In conclusion, bevacizumab-associated ICH was associated with poor prognosis in the present cohort of patients with malignant glioma. Furthermore, corpus callosum infiltration shown on magnetic resonance imaging before bevacizumab administration was suggested to be a risk factor for ICH; however, further studies on larger cohorts are required for confirmation.
Collapse
Affiliation(s)
- Yuya Hama
- Department of Neurological Surgery, School of Medicine, Wakayama Medical University, Wakayama 641-8509, Japan
| | - Takahiro Sasaki
- Department of Neurological Surgery, School of Medicine, Wakayama Medical University, Wakayama 641-8509, Japan
| | - Junya Fukai
- Department of Neurological Surgery, School of Medicine, Wakayama Medical University, Wakayama 641-8509, Japan
| | - Naoyuki Nakao
- Department of Neurological Surgery, School of Medicine, Wakayama Medical University, Wakayama 641-8509, Japan
| |
Collapse
|
17
|
Alcântara GAS, do Nascimento MC, de Miranda LBL, de Almeida BO, Lima K, Rego EM, Costa-Lotufo LV, Machado-Neto JA. Eribulin exerts multitarget antineoplastic activity in glioma cells. Pharmacol Rep 2025; 77:817-828. [PMID: 40056292 DOI: 10.1007/s43440-025-00711-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Gliomas, particularly glioblastomas, are highly aggressive cancers with rapid proliferation and poor prognosis. Current treatments have limited efficacy, highlighting the need for new therapeutic strategies. Eribulin mesylate, a synthetic macrocyclic ketone, has shown potential as an anticancer agent in several malignancies. This study investigates the cellular and molecular effects of eribulin in glioma models, focusing on its impact on cell cycle progression, apoptosis, mitochondrial function, and migration. METHODS Glioma cell lines were treated with eribulin. Cell viability was measured by MTT assay, and the cell cycle was analyzed by flow cytometry. Apoptosis was assessed through morphological changes, PARP1 cleavage, and γH2AX expression. Mitochondrial integrity and reactive oxygen species levels were evaluated by flow cytometry. Cell migration was assessed using a spheroid-based assay, and protein expression changes were analyzed by Western blotting. RESULTS Eribulin reduced cell viability, with HOG cells exhibiting the highest sensitivity. Cell cycle analysis showed G2/M phase arrest and morphological examination revealed polyploidy and apoptotic features. Mitochondrial dysfunction was observed, with decreased mitochondrial membrane potential and increased reactive oxygen species, particularly in HOG and T98G cells. Molecular analysis indicated activation of apoptotic pathways (PARP1 cleavage and γH2AX elevation) and reduced stathmin 1 expression. Eribulin also significantly reduced cell migration in HOG cells. CONCLUSION Eribulin demonstrates potent anti-glioma effects through apoptosis, mitochondrial dysfunction, and cell cycle disruption. These findings support its potential as a therapeutic option for glioblastoma treatment, warranting further investigation into its mechanisms and clinical applicability.
Collapse
Affiliation(s)
- Guilherme Augusto Sousa Alcântara
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, SP, CEP 05508-900, Brazil
| | - Mariane Cristina do Nascimento
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, SP, CEP 05508-900, Brazil
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Internal Medicine, Hematology Division, Faculdade de Medicina, University of São Paulo, São Paulo, 01246-903, Brazil
| | - Livia Bassani Lins de Miranda
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, SP, CEP 05508-900, Brazil
| | - Bruna Oliveira de Almeida
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, SP, CEP 05508-900, Brazil
| | - Keli Lima
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, SP, CEP 05508-900, Brazil
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Internal Medicine, Hematology Division, Faculdade de Medicina, University of São Paulo, São Paulo, 01246-903, Brazil
| | - Eduardo Magalhães Rego
- Laboratory of Medical Investigation in Pathogenesis and Targeted Therapy in Onco-Immuno-Hematology (LIM-31), Department of Internal Medicine, Hematology Division, Faculdade de Medicina, University of São Paulo, São Paulo, 01246-903, Brazil
| | - Leticia Veras Costa-Lotufo
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, SP, CEP 05508-900, Brazil
| | - João Agostinho Machado-Neto
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, São Paulo, SP, CEP 05508-900, Brazil.
| |
Collapse
|
18
|
Mongeon B, Craig M. Virtual Clinical Trial Reveals Significant Clinical Potential of Targeting Tumor-Associated Macrophages and Microglia to Treat Glioblastoma. CPT Pharmacometrics Syst Pharmacol 2025. [PMID: 40347051 DOI: 10.1002/psp4.70033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/10/2025] [Accepted: 03/24/2025] [Indexed: 05/12/2025] Open
Abstract
Glioblastoma is the most aggressive primary brain tumor, with a median survival of 15 months with treatment. Standard-of-care (SOC) consists of resection, radio- and chemotherapy. Clinical trials involving PD-1 inhibition with nivolumab combined with SOC failed to increase survival. A quantitative understanding of the interactions between the tumor and its immune environment that drive treatment outcomes is currently lacking. As such, we developed a mathematical model of tumor growth that considers CD8+ T cells, pro- and antitumoral tumor-associated macrophages and microglia (TAMs), SOC, and nivolumab. Using our model, we studied five TAM-targeting strategies currently under investigation for solid tumors. Our results show that PD-1 inhibition fails due to a lack of CD8+ T cell recruitment during treatment, explained by TAM-driven immunosuppressive mechanisms. Our model predicts that while reducing TAM numbers does not improve prognosis, altering their functions to counter their protumoral properties has the potential to considerably reduce post-treatment tumor burden. In particular, restoring antitumoral TAM phagocytic activity through anti-CD47 treatment in combination with SOC was predicted to nearly eradicate the tumor. By studying time-varying efficacy with the same half-life as the anti-CD47 antibody Hu5F9-G4, our model predicts that repeated dosing of anti-CD47 provides sustained control of tumor growth. We propose that targeting TAMs by enhancing their antitumoral properties is a highly promising avenue to treat glioblastoma and warrants future clinical development. Together, our results provide proof-of-concept that mechanistic mathematical modeling can uncover the mechanisms driving treatment outcomes and explore the potential of novel treatment strategies for hard-to-treat tumors like glioblastoma.
Collapse
Affiliation(s)
- Blanche Mongeon
- Sainte-Justine University Hospital Azrieli Research Centre, Montréal, Quebec, Canada
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Quebec, Canada
| | - Morgan Craig
- Sainte-Justine University Hospital Azrieli Research Centre, Montréal, Quebec, Canada
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Quebec, Canada
| |
Collapse
|
19
|
Spitzer A, Johnson KC, Nomura M, Garofano L, Nehar-Belaid D, Darnell NG, Greenwald AC, Bussema L, Oh YT, Varn FS, D'Angelo F, Gritsch S, Anderson KJ, Migliozzi S, Gonzalez Castro LN, Chowdhury T, Robine N, Reeves C, Park JB, Lipsa A, Hertel F, Golebiewska A, Niclou SP, Nusrat L, Kellet S, Das S, Moon HE, Paek SH, Bielle F, Laurenge A, Di Stefano AL, Mathon B, Picca A, Sanson M, Tanaka S, Saito N, Ashley DM, Keir ST, Ligon KL, Huse JT, Yung WKA, Lasorella A, Iavarone A, Verhaak RGW, Tirosh I, Suvà ML. Deciphering the longitudinal trajectories of glioblastoma ecosystems by integrative single-cell genomics. Nat Genet 2025:10.1038/s41588-025-02168-4. [PMID: 40346362 DOI: 10.1038/s41588-025-02168-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 03/17/2025] [Indexed: 05/11/2025]
Abstract
The evolution of isocitrate dehydrogenase (IDH)-wildtype glioblastoma (GBM) after standard-of-care therapy remains poorly understood. Here we analyzed matched primary and recurrent GBMs from 59 patients using single-nucleus RNA sequencing and bulk DNA sequencing, assessing the longitudinal evolution of the GBM ecosystem across layers of cellular and molecular heterogeneity. The most consistent change was a lower malignant cell fraction at recurrence and a reciprocal increase in glial and neuronal cell types in the tumor microenvironment (TME). The predominant malignant cell state differed between most matched pairs, but no states were exclusive or highly enriched in either time point, nor was there a consistent longitudinal trajectory across the cohort. Nevertheless, specific trajectories were enriched in subsets of patients. Changes in malignant state abundances mirrored changes in TME composition and baseline profiles, reflecting the co-evolution of the GBM ecosystem. Our study provides a blueprint of GBM's diverse longitudinal trajectories and highlights the treatment and TME modifiers that shape them.
Collapse
Affiliation(s)
- Avishay Spitzer
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- Department of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Kevin C Johnson
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Masashi Nomura
- Department of Pathology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Neurosurgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Luciano Garofano
- Department of Public Health Sciences, Division of Biostatistics and Bioinformatics, University of Miami, Miller School of Medicine, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, USA
| | | | - Noam Galili Darnell
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Alissa C Greenwald
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Lillian Bussema
- Department of Pathology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Young Taek Oh
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Frederick S Varn
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT, USA
- Institute for Systems Genomics, University of Connecticut, Storrs, CT, USA
| | - Fulvio D'Angelo
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, USA
- Department of Neurological Surgery, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Simon Gritsch
- Department of Pathology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kevin J Anderson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Simona Migliozzi
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, USA
- Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - L Nicolas Gonzalez Castro
- Department of Pathology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Tamrin Chowdhury
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Nicolas Robine
- Department of Computational Biology, New York Genome Center, New York City, NY, USA
| | - Catherine Reeves
- Department of Sequencing Operations, New York Genome Center, New York City, NY, USA
| | - Jong Bae Park
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Republic of Korea
| | - Anuja Lipsa
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Frank Hertel
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Anna Golebiewska
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Simone P Niclou
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
- University of Luxembourg Faculty of Science, Technology and Medicine, Luxembourg, Luxembourg
| | - Labeeba Nusrat
- Division of Neurosurgery, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Sorcha Kellet
- Division of Neurosurgery, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Sunit Das
- Division of Neurosurgery, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Hyo-Eun Moon
- Department of Neurosurgery, Cancer Research Institute, Hypoxia/Ischemia Disease Institute, Seoul National University, Seoul, Republic of Korea
| | - Sun Ha Paek
- Department of Neurosurgery, Cancer Research Institute, Hypoxia/Ischemia Disease Institute, Seoul National University, Seoul, Republic of Korea
- Advanced Institutes of Convergence Technology, Seoul National University, Suwon-si, Republic of Korea
| | - Franck Bielle
- Sorbonne Université, UMR S 1127, Inserm U 1127, CNRS UMR 7225, ICM-Paris Brain Institute, Equipe labellisée LNCC, Paris, France
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Neuropathology, Paris, France
| | - Alice Laurenge
- Sorbonne Université, UMR S 1127, Inserm U 1127, CNRS UMR 7225, ICM-Paris Brain Institute, Equipe labellisée LNCC, Paris, France
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Neuro-oncology, Paris, France
| | - Anna Luisa Di Stefano
- Neurology Department, Foch Hospital, Suresnes, France
- Neurosurgery Unit, Ospedali Riuniti di Livorno, Livorno, Italy
| | - Bertrand Mathon
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Neurosurgery, Paris, France
| | - Alberto Picca
- Sorbonne Université, UMR S 1127, Inserm U 1127, CNRS UMR 7225, ICM-Paris Brain Institute, Equipe labellisée LNCC, Paris, France
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Neuro-oncology, Paris, France
| | - Marc Sanson
- Sorbonne Université, UMR S 1127, Inserm U 1127, CNRS UMR 7225, ICM-Paris Brain Institute, Equipe labellisée LNCC, Paris, France
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Neuro-oncology, Paris, France
- Brain Tumor Bank Onconeurotek (ONT), AP-HP, Paris, France
| | - Shota Tanaka
- Department of Neurosurgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nobuhito Saito
- Department of Neurosurgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - David M Ashley
- Department of Neurosurgery, The Preston Robert Tisch Brain Tumor Center, Duke University, Durham, NC, USA
| | - Stephen T Keir
- Department of Neurosurgery, The Preston Robert Tisch Brain Tumor Center, Duke University, Durham, NC, USA
| | - Keith L Ligon
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jason T Huse
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - W K Alfred Yung
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anna Lasorella
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, USA.
- Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, FL, USA.
| | - Antonio Iavarone
- Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, FL, USA.
- Department of Neurological Surgery, University of Miami, Miller School of Medicine, Miami, FL, USA.
| | - Roel G W Verhaak
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA.
- Department of Neurosurgery, Amsterdam University Medical Center, Amsterdam, the Netherlands.
| | - Itay Tirosh
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| | - Mario L Suvà
- Department of Pathology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
20
|
Xu Q, Hu X, Ullah I, Lin T, Liu Y, Dong H, Zong Q, Yuan Y, Wang J. Biomimetic Hybrid PROTAC Nanovesicles Block Multiple DNA Repair Pathways to Overcome Temozolomide Resistance Against Orthotopic Glioblastoma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2504253. [PMID: 40347032 DOI: 10.1002/adma.202504253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/28/2025] [Indexed: 05/12/2025]
Abstract
Glioblastoma (GBM) remains one of the deadliest forms of cancer due to its high rates of postoperative recurrence and resistance to treatment. Temozolomide (TMZ) is the standard chemotherapy for GBM. However, the therapeutic efficacy of TMZ is significantly compromised by the activation of various intracellular DNA repair mechanisms that facilitate TMZ resistance. Herein, the upregulation of bromodomain-containing protein 4 (BRD4) expression is demonstrated to be a key contributor to TMZ resistance in GBM. To address this challenge, a biomimetic hybrid PROteolysis TArgeting Chimeras (PROTAC) liposome delivery system (M@TP) is developed. This system efficiently penetrates the blood-brain barrier (BBB) and specifically targets GBM cells through homotypic recognition. Once within TMZ-resistant GBM cells, the released PROTAC from M@TP can specifically degrade BRD4, effectively inhibiting multiple DNA repair pathways and restoring sensitivity to TMZ treatment. In vivo, studies showed that M@TP demonstrated significant efficacy in suppressing tumor growth in both TMZ-resistant and postoperative GBM, with prolonged mouse survival times. These findings highlight the potential of M@TP as a promising strategy to overcome TMZ resistance and improve therapeutic outcomes in GBM.
Collapse
Affiliation(s)
- Qing Xu
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P.R. China
| | - Xiangyu Hu
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P.R. China
| | - Ihsan Ullah
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P.R. China
| | - Taian Lin
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P.R. China
| | - Ye Liu
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P.R. China
| | - He Dong
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P.R. China
| | - Qingyu Zong
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P.R. China
| | - Youyong Yuan
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P.R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P.R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, P.R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P.R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, P. R. China
| |
Collapse
|
21
|
Uusi-Mäkelä J, Kauppinen M, Seppälä J, Jaatinen S, Ryback B, Rantapero T, Rodriguez-Martinez A, Nykter M, Rautajoki KJ. Tumor-associated long non-coding RNAs show variable expression across diffuse gliomas and effect on cell growth upon silencing in glioblastoma. Sci Rep 2025; 15:16220. [PMID: 40346283 DOI: 10.1038/s41598-025-99984-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 04/24/2025] [Indexed: 05/11/2025] Open
Abstract
Long noncoding RNAs (lncRNAs) have been recently recognized as critical components of cancer biology linked to oncogenic processes. Certain lncRNAs are known to act as oncogenes, and the disease-specific expression of many lncRNAs makes them informative biomarkers. We identified 22 uncharacterized lncRNAs from RNA-seq data of 169 glioblastoma (GBM) tumor samples sequenced by The Cancer Genome Atlas (TCGA) consortium and studied their expression in TCGA diffuse glioma cohort including also IDH-mutant astrocytomas and oligodendrogliomas as well as in normal brain samples from the Genotype-Tissue Expression cohort. All of the 22 lncRNAs were clearly upregulated in diffuse gliomas samples compared to the normal brain. Interestingly, 20 (91%) of these lncRNAs had significant expression differences between tumor grades and/or entities, and 14 (64%) were associated with overall patient survival. All 22 lncRNAs were expressed in at least one of the studied GBM cell lines and 10 (45%) were expressed in all four. When six of the lncRNAs were silenced in the SNB19 GBM cell line, the knock-down was associated with reduced growth and colony formation for three lncRNAs: TCONS_l2_00001282, lnc-GBMT-6, and lnc-NBN-1. In conclusion, the studied lncRNAs are associated with survival in patients with diffuse glioma and have functional relevance in GBM.
Collapse
Affiliation(s)
- Joonas Uusi-Mäkelä
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Maria Kauppinen
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- IT Management, Helsinki University Hospital, Helsinki, Finland
| | - Janne Seppälä
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Serafiina Jaatinen
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Birgitta Ryback
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Tommi Rantapero
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Alejandra Rodriguez-Martinez
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Matti Nykter
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Kirsi J Rautajoki
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| |
Collapse
|
22
|
Zhang D, Wang X, Xiao M, Ma S, Li S, Jia W. PLEKHA4 is transcriptionally regulated by HOXD9 and regulates glycolytic reprogramming and progression in glioblastoma via activation of the STAT3/SOCS-1 pathway. Oncogenesis 2025; 14:15. [PMID: 40346039 DOI: 10.1038/s41389-025-00559-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 04/11/2025] [Accepted: 04/29/2025] [Indexed: 05/11/2025] Open
Abstract
Recent studies have demonstrated that PLEKHA4 promotes tumor growth in some cancers, such as small-cell lung cancer, melanoma, and hepatic carcinomas; however, the underlying mechanism in glioblastoma remains ambiguous. Bioinformatic was used to analysis PLEKHA4 expression. In vitro and in vivo experiments were conducted to detect the effect of PLEKHA4 on glioblastoma cell glycolytic reprogramming and progression. GSEA was used to analyze the signal pathways related to PLEKHA4. Pharmacological methods further validated the role of activation pathways. We evaluated the effects of PLEKHA4 knockdown combined with temozolomide (TMZ) on glioblastoma cell proliferation and apoptosis in vitro and in vivo. We observed an overexpression of PLEKHA4 in GBM cell lines, resulting in enhanced cell proliferation, inhibited apoptosis, and promoted glycolysis. Mechanistically, our study demonstrated that PLEKHA4 mediates cell proliferation, apoptosis, and glycolysis via the STAT3/SOCS1 signaling pathway. Additionally, HOXD9 was predicted using Jasper, which is a transcription factor that binds to the PLEKHA4 promoter region. Knocking down PLEKHA4 combined with TMZ inhibited cell proliferation and promoted cell apoptosis in vitro and in vivo. Our results indicated that HOXD9-medicated PLEKHA4 regulates glioblastoma cell proliferation and glycolysis via activation of the STAT3/SOCS1 pathway.
Collapse
Affiliation(s)
- Dainan Zhang
- Department of Neurosurgery, Beijing Tian Hospital, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Xiaoyin Wang
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Meng Xiao
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
- Graduate School of Beijing University of Chinese Medical, Beijng, China
| | - Shunchang Ma
- Department of Neurosurgery, Beijing Tian Hospital, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Shaomin Li
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Wang Jia
- Department of Neurosurgery, Beijing Tian Hospital, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| |
Collapse
|
23
|
Su YF, Tsai TH, Kuo KL, Wu CH, Su HY, Chang WC, Huang FL, Lieu AS, Kwan AL, Loh JK, Lin CL, Tsai CY. Mitochondrial dysfunction and cell death induced by Toona sinensis leaf extracts through MEK/ERK signaling in glioblastoma cells. PLoS One 2025; 20:e0320849. [PMID: 40343958 PMCID: PMC12063860 DOI: 10.1371/journal.pone.0320849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/26/2025] [Indexed: 05/11/2025] Open
Abstract
Toona sinensis, a kind of phytochemicals in traditional Chinese medicine widely used in South-East Asia, has been recognized for its anticancer properties, particularly against various types of cancer. We aimed to evaluate the effectiveness of T. sinensis leaf extracts (TSL) specifically for glioblastoma multiforme (GBM). Gallic acid was identified as the major active component in the aqueous extracts of TSL using the HPLC system. Furthermore, it has been shown to have the ability to penetrate the blood-brain barrier. Various concentrations of TSL (10, 20, 40, and 80 μg/mL) were applied and 80 μg/mL TSL treatment significantly inhibited cell growth, proliferation, and cytotoxicity in A172 and U251 GBM cells. Flow cytometry analysis revealed cell cycle arrest at the G2/M phase and increased apoptotic cells. Furthermore, we observed mitochondrial dysfunction characterized by elevated ROS levels and reduced ATP production due to the blockade of electron transport chain (ETC) complexes. TSL treatment regulated this ROS-induced mitochondrial dysfunction. Western blotting analysis showed upregulation of Bax and Puma, along with downregulation of Bcl-2. Additionally, TSL treatment induced the cleavage of caspase-3, caspase-9, and PARP, indicating activation of the mitochondria-mediated apoptosis pathway and caspase-dependent pathway in both GBM cell lines. To investigate the involvement of the MEK/ERK pathway in TSL-induced effects, we used U0126, an inhibitor of MEK/ERK kinase. The results demonstrated that TSL treatment suppressed MEK/ERK activation, inhibiting ROS-induced mitochondrial dysfunction and promoting apoptosis. This suggests a potential therapeutic strategy targeting the MEK/ERK pathway in GBM treatment. Overall, our findings indicate that TSL treatment exerts cytotoxic effects through ROS-mediated mitochondrial dysfunction and activation of apoptotic pathways via MEK/ERK pathway in GBM cells. These insights provide valuable knowledge for potential therapeutic applications of TSL in GBM treatment.
Collapse
Affiliation(s)
- Yu-Feng Su
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Surgery, Post Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tai-Hsin Tsai
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Keng-Liang Kuo
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chieh-Hsin Wu
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hui-Yuan Su
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Wen-Chang Chang
- Department of Food Science, National Chiayi University, Chiayi, Taiwan
| | - Fu-Long Huang
- Department of Food Nutrition, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Ann-Shung Lieu
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Aij-Lie Kwan
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Joon-Khim Loh
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Lung Lin
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Cheng Yu Tsai
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Surgery, Post Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Gangshan Hospital, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
24
|
Pei D, Ma Z, Qiu Y, Wang M, Wang Z, Liu X, Zhang L, Zhang Z, Li R, Yan D. MRI-based machine learning reveals proteasome subunit PSMB8-mediated malignant glioma phenotypes through activating TGFBR1/2-SMAD2/3 axis. MOLECULAR BIOMEDICINE 2025; 6:28. [PMID: 40335825 PMCID: PMC12058589 DOI: 10.1186/s43556-025-00268-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 04/15/2025] [Accepted: 04/18/2025] [Indexed: 05/09/2025] Open
Abstract
Gliomas are the most prevalent and aggressive neoplasms of the central nervous system, representing a major challenge for effective treatment and patient prognosis. This study identifies the proteasome subunit beta type-8 (PSMB8/LMP7) as a promising prognostic biomarker for glioma. Using a multiparametric radiomic model derived from preoperative magnetic resonance imaging (MRI), we accurately predicted PSMB8 expression levels. Notably, radiomic prediction of poor prognosis was highly consistent with elevated PSMB8 expression. Our findings demonstrate that PSMB8 depletion not only suppressed glioma cell proliferation and migration but also induced apoptosis via activation of the transforming growth factor beta (TGF-β) signaling pathway. This was supported by downregulation of key receptors (TGFBR1 and TGFBR2). Furthermore, interference with PSMB8 expression impaired phosphorylation and nuclear translocation of SMAD2/3, critical mediators of TGF-β signaling. Consequently, these molecular alterations resulted in reduced tumor progression and enhanced sensitivity to temozolomide (TMZ), a standard chemotherapeutic agent. Overall, our findings highlight PSMB8's pivotal role in glioma pathophysiology and its potential as a prognostic marker. This study also demonstrates the clinical utility of MRI radiomics for preoperative risk stratification and pre-diagnosis. Targeted inhibition of PSMB8 may represent a therapeutic strategy to overcome TMZ resistance and improve glioma patient outcomes.
Collapse
Affiliation(s)
- Dongling Pei
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Zeyu Ma
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Yuning Qiu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Minkai Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Zilong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Xianzhi Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Zhenyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, China.
| | - Ran Li
- School of Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang, China.
| | - Dongming Yan
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
25
|
Kawauchi D, Narita Y. The curse of blood-brain barrier and blood-tumor barrier in malignant brain tumor treatment. Int J Clin Oncol 2025:10.1007/s10147-025-02777-3. [PMID: 40338447 DOI: 10.1007/s10147-025-02777-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Accepted: 04/24/2025] [Indexed: 05/09/2025]
Abstract
The blood-brain barrier (BBB) is crucial for brain homeostasis but is a major obstacle in delivering anticancer drugs to brain tumors. However, this perspective requires re-evaluation, particularly for malignant brain tumors, such as gliomas and brain metastases. In these aggressive tumors, the BBB undergoes significant alterations, leading to the formation of a more permeable blood-tumor barrier. While this increased permeability allows better drug penetration, heterogeneity in blood-tumor barrier (BTB) integrity across different tumor regions remains a challenge. Additionally, the main challenge in treating brain tumors lies not in BBB penetration but in the lack of effective drugs. Conventional chemotherapies, including temozolomide and nitrosourea agents, have shown limited efficacy, and resistance mechanisms often reduce their long-term benefits. The "BBB curse" has often been blamed for the slow progress in drug development. However, emerging evidence suggests that even larger-molecule therapies, such as antibody-drug conjugates, can successfully target brain tumors. This review aims to critically reassess the roles of the BBB and BTB in brain tumor therapy, highlighting their impact on drug delivery and evaluating the current landscape of chemotherapeutic strategies. Furthermore, it explores new approaches to overcome treatment limitations, emphasizing the need for personalized and targeted therapeutic strategies.
Collapse
Affiliation(s)
- Daisuke Kawauchi
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Chuo City, Japan
| | - Yoshitaka Narita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Chuo City, Japan.
| |
Collapse
|
26
|
Bennett J, Levine AB, Nobre L, Negm L, Chung J, Fang K, Johnson M, Komosa M, Krumholtz S, Nunes NM, Rana M, Ryall S, Sheth J, Siddaway R, Bale TA, Bouffet E, Cusimano MD, Das S, Detsky J, Dirks P, Karajannis MA, Kongkham P, Giantini-Larsen A, Li BK, Lim-Fat MJ, Lin AL, Mason WP, Miller A, Perry JR, Sahgal A, Sait SF, Tsang DS, Zadeh G, Laperriere N, Nguyen L, Gao A, Keith J, Munoz DG, Tabori U, Hawkins C. A population-based analysis of the molecular landscape of glioma in adolescents and young adults reveals insights into gliomagenesis. NATURE CANCER 2025:10.1038/s43018-025-00962-x. [PMID: 40335748 DOI: 10.1038/s43018-025-00962-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 03/28/2025] [Indexed: 05/09/2025]
Abstract
Gliomas are a major cause of cancer-related deaths in adolescents and young adults (AYAs; ages 15-39 years). Different molecular alterations drive gliomas in children and adults, leading to distinct biology and clinical consequences, but the implications of pediatric- versus adult-type alterations in AYAs are unknown. Our population-based analysis of 1,456 clinically and molecularly characterized gliomas in patients aged 0-39 years addresses this gap. Pediatric-type alterations were found in 31% of AYA gliomas and conferred superior outcomes compared to adult-type alterations. AYA low-grade gliomas with specific RAS-MAPK alterations exhibited senescence, tended to arise in different locations and were associated with superior outcomes compared to gliomas in children, suggesting different cellular origins. Hemispheric IDH-mutant, BRAF p.V600E and FGFR-altered gliomas were associated with the risk of malignant transformation, having worse outcomes with increased age. These insights into gliomagenesis may provide a rationale for earlier intervention for certain tumors to disrupt the typical behavior, leading to improved outcomes.
Collapse
Affiliation(s)
- Julie Bennett
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada.
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada.
| | - Adrian B Levine
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Liana Nobre
- Division of Hematology/Oncology (iHOPE), Department of Pediatrics, Stollery Children's Hospital, University of Alberta, Edmonton, Alberta, Canada
| | - Logine Negm
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jiil Chung
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Karen Fang
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Monique Johnson
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Martin Komosa
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Stacey Krumholtz
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Nuno Miguel Nunes
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Mansuba Rana
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Scott Ryall
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Javal Sheth
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Robert Siddaway
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Tejus A Bale
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eric Bouffet
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michael D Cusimano
- Division of Neurosurgery, Unity Health, Toronto, Ontario, Canada
- Division of Neurosurgery, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Neurosurgery, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Sunit Das
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Neurosurgery, Unity Health, Toronto, Ontario, Canada
| | - Jay Detsky
- Department of Radiation Oncology, Sunnybrook Health Science Centre, Toronto, Ontario, Canada
| | - Peter Dirks
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Neurosurgery, The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Paul Kongkham
- Department of Neurosurgery, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | | | - Bryan Kincheon Li
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Division of Pediatric Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mary Jane Lim-Fat
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Andrew L Lin
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Warren P Mason
- Department of Medicine, Princess Margaret Cancer Centre, University Health Network and University of Toronto, Toronto, Ontario, Canada
| | - Alexandra Miller
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - James R Perry
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Arjun Sahgal
- Department of Radiation Oncology, Sunnybrook Health Science Centre, Toronto, Ontario, Canada
| | - Sameer Farouk Sait
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Derek S Tsang
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Gelareh Zadeh
- Department of Neurosurgery, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Normand Laperriere
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Lananh Nguyen
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine, Unity Health, Toronto, Ontario, Canada
| | - Andrew Gao
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Laboratory Medicine Program, University Health Network, Toronto, Ontario, Canada
| | - Julia Keith
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - David G Munoz
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine, Unity Health, Toronto, Ontario, Canada
| | - Uri Tabori
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Cynthia Hawkins
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
27
|
Ivanova YI, Nunes AC, Cruz V, Selting K, Harley BAC. Radiation Damage to a Three-Dimensional Hydrogel Model of the Brain Perivascular Niche. Tissue Eng Part C Methods 2025. [PMID: 40329812 DOI: 10.1089/ten.tec.2025.0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025] Open
Abstract
Glioblastoma (GBM) is a highly aggressive and recurrent brain cancer characterized by diffuse metastasis at the tumor margins. Radiation therapy is a standard component of current treatment and offers potential for improved patient outcomes. While radiation therapy targets GBM cells in the tumor margins, it may also significantly damage adjacent noncancerous tissues, leading to reduced quality of life and potentially creating a tumor-supportive microenvironment. The perivascular niche (PVN) in the tumor margins is believed to play a significant role in regulating the glioblastoma stem cell subpopulation as well as serving as a site for cancer recurrence and migration. Understanding the impact of radiation on the PVN can better inform radiation schemes and improve our understanding of GBM recurrence, but is difficult in vivo. Here, we adapt a previously developed three-dimensional hydrogel model of the brain PVN to investigate the impact of radiation dosage and delivery rate on PVN properties in vitro. Effects of radiation on vessel architecture can be measured in this hydrogel-based model, suggesting an approach that can provide insight into the effects of radiation on a shorter time scale relative to in vivo experiments.
Collapse
Affiliation(s)
- Yoanna I Ivanova
- Department of Bioengineering, University of Illinois, Urbana, Illinois, USA
| | - Alison C Nunes
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Val Cruz
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Kimberly Selting
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Brendan A C Harley
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
28
|
Zhang L, Yang J, Zhou Z, Ren Y, Chen B, Tang A, Zhang K, Li C, Zhou H, Fung KM, Xu C, Kang C, Battiste JD, Bronze MS, Houchen CW, Liu Z, Dunn IF, Cavenee WK, Li M. A zinc transporter drives glioblastoma progression via extracellular vesicles-reprogrammed microglial plasticity. Proc Natl Acad Sci U S A 2025; 122:e2427073122. [PMID: 40305049 DOI: 10.1073/pnas.2427073122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 03/22/2025] [Indexed: 05/02/2025] Open
Abstract
Glioblastoma (GBM) is the most aggressive form of brain cancer, with limited therapeutic options. While microglia contribute to GBM progression, the mechanisms by which they foster a protumorigenic immune environment remain poorly understood. We identify the zinc transporter Zrt- And Irt-Like Protein 4 (ZIP4) as a pivotal regulator of the GBM immune landscape. In orthotopic mouse models, ZIP4 drives tumor growth and behavioral changes. Mechanistically, ZIP4 modulates microglial plasticity through tumor-derived extracellular vesicles carrying triggering receptor expressed on myeloid cells-1 (TREM1), a process regulated by the zinc-dependent transcription factor Zinc Finger E-box Binding Homeobox 1 in GBM cells. TREM1 enhances microglial plasticity through the spleen associated tyrosine kinase-Pyruvate dehydrogenase kinase-signal transducer and activator of transcription 3 (SYK-PDK-STAT3) signaling axis, ultimately promoting an immune environment favorable to tumor progression. ZIP4 depletion or TREM1 inhibition attenuates tumor growth and behavioral effects in vivo by disrupting the tumor-microglia interaction. These findings establish ZIP4 as a key modulator of the GBM immune landscape and suggest a promising therapeutic target to counteract microglia-mediated tumor progression.
Collapse
Affiliation(s)
- Liyang Zhang
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jingxuan Yang
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Zhijun Zhou
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Yu Ren
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Bo Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Anliu Tang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Kailiang Zhang
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Chuntao Li
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Hongshu Zhou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Kar-Ming Fung
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Chao Xu
- Department of Biostatistics and Epidemiology, College of Public Health, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Chunsheng Kang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin 300052, China
| | - James D Battiste
- Department of Neurosurgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Michael S Bronze
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Courtney W Houchen
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Ian F Dunn
- Department of Neurosurgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Webster K Cavenee
- Department of Medicine, University of California at San Diego, San Diego, CA 92093
| | - Min Li
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| |
Collapse
|
29
|
Song KW, Lim M, Monje M. Complex neural-immune interactions shape glioma immunotherapy. Immunity 2025:S1074-7613(25)00180-3. [PMID: 40324379 DOI: 10.1016/j.immuni.2025.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 05/07/2025]
Abstract
Rich neural-immune interactions in the central nervous system (CNS) shape its function and create a unique immunological microenvironment for immunotherapy in CNS malignancies. Far from the now-debunked concept of CNS "immune privilege," it is now understood that unique immunological niches and constant immune surveillance of the brain contribute in multifaceted ways to brain health and robustly influence immunotherapy approaches for CNS cancers. Challenges include immune-suppressive and neurotoxicity-promoting crosstalk between brain, immune, and tumor cells. Developing effective immunotherapies for cancers of the nervous system will require a deeper understanding of these neural-immune-malignant cell interactions. Here, we review progress and challenges in immunotherapy for gliomas of the brain and spinal cord in light of these unique neural-immune interactions and highlight future work needed to optimize promising immunotherapies for gliomas.
Collapse
Affiliation(s)
- Kun-Wei Song
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, USA
| | - Michael Lim
- Department of Neurosurgery, Stanford University, Palo Alto, CA, USA
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA, USA; Department of Neurosurgery, Stanford University, Palo Alto, CA, USA; Howard Hughes Medical Institute, Stanford University, Palo Alto, CA, USA.
| |
Collapse
|
30
|
Zabihi A. The role of biological macromolecules in the regulation of angiogenesis in glioblastoma: Focus on vascular growth factors, integrins, and extracellular matrix proteins. Int J Biol Macromol 2025; 311:143838. [PMID: 40319984 DOI: 10.1016/j.ijbiomac.2025.143838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
Glioblastoma, classified as a grade 4 brain tumor, accounts for approximately half of all malignant central nervous system cancers. Despite extensive research and aggressive treatment modalities, much about this disease remains elusive. The proliferation of blood vessels within glioblastoma tumors significantly contributes to their invasive nature, primarily due to the influence of vascular endothelial growth factor-A (VEGF-A). As a result, the past decade has seen a concentrated effort to explore angiogenesis, especially the VEGF signaling pathway, as a therapeutic target for glioblastoma. This investigation led to the FDA approval of bevacizumab, a monoclonal antibody against VEGF-A, for the treatment of recurrent glioblastoma. However, despite promising clinical trials and theoretical research, bevacizumab has not significantly improved patient survival rates. Furthermore, other anti-angiogenic agents targeting the VEGF signaling pathway have shown limited efficacy. This suggests the existence of multiple alternative angiogenic pathways that facilitate vascularization, even when VEGF signaling is inhibited. In this study, we aim to assess the current landscape of anti-angiogenic agents, explore potential resistance mechanisms to such therapies, and suggest strategies to improve the effectiveness of these therapeutic interventions. Our goal is to provide a comprehensive understanding of the limitations of current treatments and to identify new avenues for enhancing therapeutic outcomes in glioblastoma patients.
Collapse
Affiliation(s)
- Abbas Zabihi
- Department of Biology, Faculty of Basic Sciences, Islamic Azad University Rasht Branch, Rasht, Iran.
| |
Collapse
|
31
|
O'Keeffe P, Nouri Y, Saw HS, Moore Z, Baldwin TM, Olechnowicz SWZ, Jabbari JS, Squire DM, Leslie S, Wang C, You Y, Ritchie ME, Cross RS, Jenkins MR, Audiger C, Naik SH, Whittle JR, Freytag S, Best SA, Hickey PF, Amann-Zalcenstein D, Bowden R, Brown DV. TIRE-seq simplifies transcriptomics via integrated RNA capture and library preparation. Sci Rep 2025; 15:15385. [PMID: 40316593 PMCID: PMC12048512 DOI: 10.1038/s41598-025-98282-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 04/10/2025] [Indexed: 05/04/2025] Open
Abstract
RNA sequencing (RNA-seq) is widely used in biomedical research, advancing our understanding of gene expression across biological systems. Traditional methods require upstream RNA extraction from biological inputs, adding time and expense to workflows. We developed TIRE-seq (Turbocapture Integrated RNA Expression Sequencing) to address these challenges. TIRE-seq integrates mRNA purification directly into library preparation, eliminating the need for a separate extraction step. This streamlined approach reduces turnaround time, minimizes sample loss, and improves data quality. A comparative study with the widely used Prime-seq protocol demonstrates TIRE-seq's superior sequencing efficiency with crude cell lysates as inputs. TIRE-seq's utility was demonstrated across three biological applications. It captured transcriptional changes in stimulated human T cells, revealing activation-associated gene expression profiles. It also identified key genes driving murine dendritic cell differentiation, providing insights into lineage commitment. Lastly, TIRE-seq analyzed the dose-response and time-course effects of temozolomide on patient-derived neurospheres, identifying differentially expressed genes and enriched pathways linked to the drug's mechanism of action. With its simplified workflow and high sequencing efficiency, TIRE-seq offers a cost-effective solution for large-scale gene expression studies across diverse biological systems.
Collapse
Affiliation(s)
- Piper O'Keeffe
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade VIC, Melbourne, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Yasmin Nouri
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade VIC, Melbourne, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Hui Shi Saw
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade VIC, Melbourne, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Zachery Moore
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade VIC, Melbourne, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Tracey M Baldwin
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade VIC, Melbourne, VIC, 3052, Australia
| | - Sam W Z Olechnowicz
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade VIC, Melbourne, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jafar S Jabbari
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade VIC, Melbourne, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - David McG Squire
- Melbourne Integrative Genomics, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Stephen Leslie
- Melbourne Integrative Genomics, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Changqing Wang
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade VIC, Melbourne, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Yupei You
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade VIC, Melbourne, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Matthew E Ritchie
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade VIC, Melbourne, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Ryan S Cross
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade VIC, Melbourne, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Misty R Jenkins
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade VIC, Melbourne, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Cindy Audiger
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade VIC, Melbourne, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Shalin H Naik
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade VIC, Melbourne, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - James R Whittle
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade VIC, Melbourne, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
- Peter MacCallum Cancer Centre, Parkville, VIC, 3010, Australia
| | - Saskia Freytag
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade VIC, Melbourne, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Sarah A Best
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade VIC, Melbourne, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Peter F Hickey
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade VIC, Melbourne, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Daniela Amann-Zalcenstein
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade VIC, Melbourne, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Rory Bowden
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade VIC, Melbourne, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Daniel V Brown
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade VIC, Melbourne, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
32
|
Zhao C, Guo Y, Chen Y, Shang G, Song D, Wang J, Yang J, Zhang H. RETRACTED: Zinc finger Protein207 orchestrates glioma migration through regulation of epithelial-mesenchymal transition. ENVIRONMENTAL TOXICOLOGY 2025; 40:E59-E73. [PMID: 38591780 DOI: 10.1002/tox.24271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/15/2024] [Accepted: 03/24/2024] [Indexed: 04/10/2024]
Abstract
BACKGROUND Glioma represents the predominant primary malignant brain tumor. For several years, molecular profiling has been instrumental in the management and therapeutic stratification of glioma, providing a deeper understanding of its biological complexity. Accumulating evidence unveils the putative involvement of zinc finger proteins (ZNFs) in cancer. This study aimed to elucidate the role and significance of ZNF207 in glioma. METHODS Utilizing online data such as The Cancer Genome Atlas (TCGA), the Chinese Glioma Genome Atlas (CGGA), the Genotype-Tissue Expression (GTEx) project, the Clinical Proteomic Tumor Analysis Consortium (CPTAC), and the Human Protein Atlas (HPA) databases, in conjunction with bioinformatics methodologies including GO, KEGG, GSEA, CIBERSORT immune cell infiltration estimation, and protein-protein interaction (PPI) analysis, enabled a comprehensive exploration of ZNF207's involvement in gliomagenesis. Immunohistochemistry and RT-PCR techniques were employed to validate the expression level of ZNF207 in glioma samples. Subsequently, the biological effects of ZNF207 on glioma cells were explored through in vitro assays. RESULTS Our results demonstrate elevated expression of ZNF207 in gliomas, correlating with unfavorable patient outcomes. Stratification analyses were used to delineate the prognostic efficacy of ZNF207 in glioma with different clinicopathological characteristics. Immunocorrelation analysis revealed a significant association between ZNF207 expression and the infiltration levels of T helper cells, macrophages, and natural killer (NK) cells. Utilizing ZNF207 expression and clinical features, we constructed an OS prediction model and displayed well discrimination with a C-index of 0.861. Moreover, the strategic silencing of ZNF207 attenuated glioma cell advancement, evidenced by diminished cellular proliferation, weakened cell tumorigenesis, augmented apoptotic activity, and curtailed migratory capacity alongside the inhibition of the epithelial-mesenchymal transition (EMT) pathway. CONCLUSIONS ZNF207 may identify as a prospective biomarker and therapeutic candidate for glioma prevention, providing valuable insights into understanding glioma pathogenesis and treatment strategies.
Collapse
Affiliation(s)
- Chao Zhao
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Yuduo Guo
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Yujia Chen
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Guanjie Shang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Dixiang Song
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Jun Wang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Jingjing Yang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Hongwei Zhang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
33
|
Mansoorian S, Schmidt M, Weissmann T, Delev D, Heiland DH, Coras R, Stritzelberger J, Saake M, Höfler D, Schubert P, Schmitter C, Lettmaier S, Filimonova I, Frey B, Gaipl US, Distel LV, Semrau S, Bert C, Eze C, Schönecker S, Belka C, Blümcke I, Uder M, Schnell O, Dörfler A, Fietkau R, Putz F. Reirradiation for recurrent glioblastoma: the significance of the residual tumor volume. J Neurooncol 2025:10.1007/s11060-025-05042-9. [PMID: 40310485 DOI: 10.1007/s11060-025-05042-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025]
Abstract
PURPOSE Recurrent glioblastoma has a poor prognosis, and its optimal management remains unclear. Reirradiation (re-RT) is a promising treatment option, but long-term outcomes and optimal patient selection criteria are not well established. METHODS This study analyzed 71 patients with recurrent CNS WHO grade 4, IDHwt glioblastoma (GBM) who underwent re-RT at the University of Erlangen-Nuremberg between January 2009 and June 2019. Imaging follow-ups were conducted every 3 months. Progression-free survival (PFS) was defined using RANO criteria. Outcomes, feasibility, and toxicity of re-RT were evaluated. Contrast-enhancing tumor volume was measured using a deep learning auto-segmentation pipeline with expert validation and jointly evaluated with clinical and molecular-pathologic factors. RESULTS Most patients were prescribed conventionally fractionated re-RT (84.5%) with 45 Gy in 1.8 Gy fractions, combined with temozolomide (TMZ, 49.3%) or lomustine (CCNU, 12.7%). Re-RT was completed as planned in 94.4% of patients. After a median follow-up of 73.8 months, 88.7% of patients had died. The median overall survival was 9.6 months, and the median progression-free survival was 5.3 months. Multivariate analysis identified residual contrast-enhancing tumor volume at re-RT (HR 1.040 per cm3, p < 0.001) as the single dominant predictor of overall survival. CONCLUSION Conventional fractionated re-RT is a feasible and effective treatment for recurrent high-grade glioma. The significant prognostic impact of residual tumor volume highlights the importance of combining maximum-safe resection with re-RT for improved outcomes.
Collapse
Affiliation(s)
- Sina Mansoorian
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitaetsstraße 27, 91054, Erlangen, Germany
- Department of Radiation Oncology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Manuel Schmidt
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- Department of Neuroradiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Weissmann
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitaetsstraße 27, 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Daniel Delev
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- Department of Neurosurgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Dieter Henrik Heiland
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- Department of Neurosurgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Roland Coras
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- Department of Neuropathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jenny Stritzelberger
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- Epilepsy Center, Department of Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Marc Saake
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Daniel Höfler
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitaetsstraße 27, 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Philipp Schubert
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitaetsstraße 27, 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Charlotte Schmitter
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitaetsstraße 27, 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Sebastian Lettmaier
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitaetsstraße 27, 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Irina Filimonova
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitaetsstraße 27, 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Benjamin Frey
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitaetsstraße 27, 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Udo S Gaipl
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitaetsstraße 27, 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Luitpold V Distel
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitaetsstraße 27, 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Sabine Semrau
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitaetsstraße 27, 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Christoph Bert
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitaetsstraße 27, 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Chukwuka Eze
- Department of Radiation Oncology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Stephan Schönecker
- Department of Radiation Oncology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Ingmar Blümcke
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- Department of Neuropathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Uder
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Oliver Schnell
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- Department of Neurosurgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Arnd Dörfler
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- Department of Neuroradiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitaetsstraße 27, 91054, Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Florian Putz
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitaetsstraße 27, 91054, Erlangen, Germany.
- Bavarian Cancer Research Center (BZKF), Munich, Germany.
| |
Collapse
|
34
|
De Silva MI, Gan HK, Bardy C. Repurposing trifluoperazine for glioblastoma treatment. Trends Pharmacol Sci 2025; 46:392-406. [PMID: 40300936 DOI: 10.1016/j.tips.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/16/2025] [Accepted: 03/16/2025] [Indexed: 05/01/2025]
Abstract
Glioblastoma (GBM) remains a therapeutic challenge due to its heterogeneity and plasticity, which drive treatment resistance, especially when compounded by interactions with the brain microenvironment. Recent preclinical evidence indicates that trifluoperazine (TFP) inhibits treatment-induced malignant reprogramming of tumour cells, potentially helping to reduce tumour plasticity. TFP targets calmodulin, dopamine receptors, and stress-responsive proteins (nuclear protein 1, NUPR1). Through these mechanisms, TFP has been shown to reduce tumour growth, sensitise tumours to chemoradiotherapy, and prolong survival in xenograft animal models. The clinical safety profile of TFP is well known from its use as an antipsychotic, and recent preclinical evidence further indicates that TFP has low toxicity to healthy neurons and glia despite transient functional effects on dopamine receptors. This Opinion explores TFP mechanisms of action and clinical activity to assess its suitability as a repurposed therapeutic option for GBM.
Collapse
Affiliation(s)
- Manam Inushi De Silva
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia; South Australian Health and Medical Research Institute (SAHMRI), Laboratory for Human Neurophysiology and Genetics, Adelaide, SA, Australia
| | - Hui K Gan
- Cancer Therapies and Biology Group, Centre of Research Excellence in Brain Tumours, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, VIC, Australia; La Trobe University School of Cancer Medicine, and Department of Medicine, University of Melbourne, Heidelberg, Melbourne, VIC, Australia
| | - Cedric Bardy
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia; South Australian Health and Medical Research Institute (SAHMRI), Laboratory for Human Neurophysiology and Genetics, Adelaide, SA, Australia.
| |
Collapse
|
35
|
Vejzovic D, Kubin A, Fechter K, Karner C, Hartmann J, Ackerbauer T, Radović B, Ritter G, Üçal M, Ropele S, Ali KM, Mischkulnig M, Leoni M, Liegl-Atzwanger B, Wightman L, Rinner B. Glioblastoma targeting by water-soluble hypericin derivate HHL-PVP and photodynamic tumour killing. Biomed Pharmacother 2025; 186:118041. [PMID: 40228364 DOI: 10.1016/j.biopha.2025.118041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/12/2025] [Accepted: 04/03/2025] [Indexed: 04/16/2025] Open
Abstract
Hypericin, a tumour-selective photosensitizer, has shown potential in cancer therapy, but its poor water solubility has limited clinical use. To address this, we developed a water-soluble variant called high hypericin-loaded polyvinylpyrrolidone (HHL-PVP) to enhance hypericin's applicability, particularly for treating glioblastoma, a typically terminal disease. We tested HHL-PVP in both in vitro and in vivo models, first confirming its fluorescent properties in the lab and then assessing its efficacy in more complex animal models. Using subcutaneous and orthotopic tumour mouse models, we combined HHL-PVP administration with fluorescence-guided surgery and photodynamic therapy (PDT) to target residual tumour cells. Histological analysis of both healthy and tumour tissue showed HHL-PVP's over 97 % sensitivity and 100 % specificity in distinguishing tumour tissue. In subcutaneous glioblastoma models, significant tumour necrosis and remission occurred after HHL-PVP administration and a 20-minute white light application through the skin. These results highlight HHL-PVP's effectiveness in targeting and eradicating glioblastoma cells. Our findings provide strong evidence that HHL-PVP is a promising therapeutic option for glioblastoma, with its high sensitivity, specificity, and potential for tumour remission through PDT. This approach warrants further investigation in clinical trials and could improve outcomes for a disease that has been difficult to treat.
Collapse
Affiliation(s)
- Djenana Vejzovic
- Division of Biomedical Research, Medical University of Graz, Graz, Austria
| | | | - Karoline Fechter
- Division of Biomedical Research, Medical University of Graz, Graz, Austria
| | - Christina Karner
- Division of Biomedical Research, Medical University of Graz, Graz, Austria
| | | | | | - Branislav Radović
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | - Gerald Ritter
- Division of Biomedical Research, Medical University of Graz, Graz, Austria
| | - Muammer Üçal
- Department of Neurosurgery, Research Unit for Experimental Neurotraumatology, Medical University of Graz, Graz, Austria; Department of Neurology, Medical University of Graz, Graz, Austria; BioTechMed Graz, Graz, Austria
| | - Stefan Ropele
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Kariem Mahdy Ali
- Department of Neurosurgery, Research Unit for Experimental Neurotraumatology, Medical University of Graz, Graz, Austria
| | | | - Marlene Leoni
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | | | | | - Beate Rinner
- Division of Biomedical Research, Medical University of Graz, Graz, Austria; BioTechMed Graz, Graz, Austria.
| |
Collapse
|
36
|
Lien JC, Hsu SY, Chueh FS, Ma YS, Chu YL, Chou YC, Lai KC, Chen JC, Huang YP, Wu RSC. Newly Synthesized PW06 Induced Cell Apoptosis in Human Glioblastoma Multiforme GBM 8401 Cells Through Caspase- and Mitochondria-Dependent Pathways. J Biochem Mol Toxicol 2025; 39:e70264. [PMID: 40258141 DOI: 10.1002/jbt.70264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 02/08/2025] [Accepted: 04/04/2025] [Indexed: 04/23/2025]
Abstract
Glioblastoma multiforme (GBM) is the most common, aggressive, and dangerous lethal tumor in the brain, which develops in adults. Currently, the efficiency of chemotherapy treatment for GBM patients is still unsatisfactory. PW06 was synthesized by Dr. Lien's laboratory (China Medical University, Taichung, Taiwan), and it was demonstrated to induce cancer cell apoptosis in human pancreatic carcinoma MIA PaCa-2 cells. However, the anti-cancer activities of PW06 on human GBM cancer cells are not reported. Thus, herein, PW06 was investigated on the anticancer activity on human glioblastoma multiforme GBM 8401 cells. Both PI exclusion and Annexin V/PI double staining methods were conducted for investing cell viability and apoptosis in GBM 8401 cells, respectively; they were analyzed with flow cytometer assay. Results showed that PW06 decreased total viable cell number with the process of cell apoptosis in GBM 8401 cells. Both productions of reactive oxygen species (ROS) and Ca2+, affect mitochondria membrane potential (ΔΨm) levels, and activities of caspase-3, -8, and -9 in GBM 8401 cells after exposure with PW06 were assayed by flow cytometer. Results showed that PW06 promoted ROS production and Ca2+ release from ER but lowered the levels of ΔΨm, and it also induced higher activities in caspase-3, -8, and -9 in GBM 8401 cells. Evaluation of protein expressions associated with apoptosis in GBM 8401 cells after being incubated with PW06 were conducted by Western blot analysis. Results show that PW06 increased GADD153, BiP, ATF-6α, ATF-6β, eIF2α, eIF2αpSer51, CHOP, and caspase-4, and they are associated with ER stress-associated protein expression. However, it induced higher pro-apoptotic proteins (Bax and Bad) expression and inhibited anti-apoptotic proteins (Bcl-2, Bcl-xl, and Mcl-1) expression, even promoting higher cleaved caspase-8, -9, and -3 protein expression and increased EndoG and AIF in GBM 8401 cells. Collectively, it may suggest PW06 exits anti-GBM activity to process cell apoptosis in the human GBM 8401 cells in vitro.
Collapse
Affiliation(s)
- Jin-Cherng Lien
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Sheng-Yao Hsu
- Department of Ophthalmology, Kaohsiung Show Chwan Memorial Hospital, Tainan, Taiwan
- Department of Optometry, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Fu-Shin Chueh
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung, Taiwan
| | - Yi-Shih Ma
- School of Chinese Medicine for Post-Baccalaureate, College of Medicine, I-Shou University, Kaohsiung, Taiwan
- Department of Chinese Medicine, E-Da Cancer Hospital, Kaohsiung, Taiwan
| | - Yung-Lin Chu
- Department of Food Science, College of Agriculture, National Pingtung University of Science Technology, Pingtung, Taiwan
| | - Yu-Cheng Chou
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Kuang-Chi Lai
- Department of Surgery, School of Medicine, China Medical University, Taichung, Taiwan
| | - Jaw-Chyun Chen
- Department of Medicinal Botanicals and Foods on Health Applications, Da-Yeh University, Changhua, Taiwan
| | - Yi-Ping Huang
- Department of Physiology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Rick Sai-Chuen Wu
- Department of Anesthesiology, China Medical University Hospital, Taichung, Taiwan
- Department of Anesthesiology, China Medical University, Taichung, Taiwan
| |
Collapse
|
37
|
Lucas Calduch A, Macià Garau M, Villà Freixa S, García Expósito N, Modolell Farré I, Majós Torró C, Pons Escoda A, Mesía Barroso C, Vilariño Quintela N, Rosselló Gómez A, Plans Ahicart G, Martínez García M, Esteve Gómez A, Bruna Escuer J. Salvage reirradiation for recurrent glioblastoma: a retrospective case series analysis. Clin Transl Oncol 2025; 27:2104-2112. [PMID: 39388047 DOI: 10.1007/s12094-024-03750-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024]
Abstract
PURPOSE To assess the clinical outcome of patients with recurrent glioblastoma treated with salvage reirradiation. METHODS Between 2005 and 2022, data from adult patients with glioblastoma treated with surgery and radio-chemotherapy Stupp regimen who developed a local in-field relapse and received stereotactic radiotherapy (SRT) were retrospectively reviewed. RESULTS The study population included 44 patients with recurrent glioblastoma (median of 9.5 months after the first radiotherapy). Reirradiation alone was given to 47.7% of patients. The median maximum diameter of the recurrence was 13.5 mm. The most common SRT regimen (52.3%) was 35 Gy in 10 fractions. Acute toxicity was mild, with transient worsening of previous neurological symptoms in only 15% of patients. After a median follow-up of 15 months, 40% presented radiological response, but a remarkable number of early distant progressions were recorded (32.5%). The median time to progression was 4.8 months, being the dose, the scheme, the size of the recurrence or the strategy (exclusive RT vs. combined) unrelated factors. The median overall survival (OS) was 14.9 months. Karnofsky index < 70 and the size of the recurrence (maximum diameter < 25 mm) were significant factors associated with OS. Radiological changes after reirradiation were commonly seen (> 50% of patients) hindering the response assessment. CONCLUSIONS Reirradiation is a feasible and safe therapeutic option to treat localized glioblastoma recurrences, able to control the disease for a few months in selected patients, especially those with good functional status and small lesions. Hypofractionated schemes provided a suitable toxicity profile. Radiological changes were common.
Collapse
Affiliation(s)
- Anna Lucas Calduch
- Radiation Oncology Service, Institut Català d'Oncologia (ICO)-Hospital Duran I Reynals, L'Hospitalet de Llobregat, Avda. Gran Via de L'Hospitalet 199-203, 08908, Barcelona, Spain.
| | - Miquel Macià Garau
- Radiation Oncology Service, Institut Català d'Oncologia (ICO)-Hospital Duran I Reynals, L'Hospitalet de Llobregat, Avda. Gran Via de L'Hospitalet 199-203, 08908, Barcelona, Spain
| | | | - Nagore García Expósito
- Radiation Oncology Service, Institut Català d'Oncologia (ICO)-Hospital Duran I Reynals, L'Hospitalet de Llobregat, Avda. Gran Via de L'Hospitalet 199-203, 08908, Barcelona, Spain
| | - Ignasi Modolell Farré
- Medical Physics, Institut Català de'Oncologia (ICO), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Carles Majós Torró
- Institut de Diagnòstic per la Imatge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Albert Pons Escoda
- Institut de Diagnòstic per la Imatge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Carlos Mesía Barroso
- Medical Oncology, Institut Català de'Oncologia (ICO)- L'Hospitalet de Llobregat, Barcelona, Spain
| | - Noelia Vilariño Quintela
- Medical Oncology, Institut Català de'Oncologia (ICO)- L'Hospitalet de Llobregat, Barcelona, Spain
| | - Aleix Rosselló Gómez
- Neurosurgery, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Gerard Plans Ahicart
- Neurosurgery, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Anna Esteve Gómez
- Badalona Applied Research Group in Oncology (B-ARGO), Badalona, Barcelona, Spain
| | - Jordi Bruna Escuer
- Neurology, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
38
|
Stummer W, Gerwing M, Bilgin SS, Thomas C, Villanueva-Meyer J, Agarwal V, Stögbauer L, Schroeteler J, Müther M. Sonodynamic therapy with a single neoadjuvant, diffuse delivery of low-intensity ultrasound with 5-ALA in treatment naïve glioblastoma results in tumor-specific cytotoxic edema and increased apoptosis. J Neurooncol 2025; 172:687-693. [PMID: 39904876 PMCID: PMC11968568 DOI: 10.1007/s11060-025-04957-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
PURPOSE Sonodynamic therapy, which combines a tumor cell-selective sonosensitizer with ultrasound, is gaining attention as a promising new treatment approach for glioblastoma. The objective of this case study is to report on the first applications of 5-aminolevulinic acid (5-ALA) in combination with low-intensity, non-targeted ultrasound as neo-adjuvant treatment in therapy naïve glioblastoma. METHODS Three patients with therapy naïve newly diagnosed glioblastoma were treated once before cytoreductive surgery with 5-ALA in combination with hemispheric, low-intensity, non-targeted ultrasound, assuming cell death to be triggered by non-ablative activation of 5-ALA-induced, tumor selective porphyrins. RESULTS No adverse effects were noted. Post-procedural MRI indicated a decrease in apparent diffusion coefficient values in tumors, suggesting cytotoxic effects. Relative cerebral blood volumes and leakage were increased for two patients with available perfusion imaging. Tissue obtained during surgery suggested increased cleaved-caspase III expression, a marker of apoptosis. CONCLUSION We saw an immediate marked imaging response indicating cytotoxic edema and indications of a histopathology response from just a single treatment. Correlation to clinical outcomes and extension of overall survival remains to be seen. A Phase 1 safety study has been submitted for regulatory approval.
Collapse
Affiliation(s)
- Walter Stummer
- Department of Neurosurgery, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.
| | - Mirjam Gerwing
- Department of Radiology, University Hospital Münster, Münster, Germany
| | | | - Christian Thomas
- Institute of Neuropathology, University Münster, Münster, Germany
| | | | - Vijay Agarwal
- Montefiore Health Center, Department of Neurological Surgery, New York, NY, USA
| | - Louise Stögbauer
- Department of Neurosurgery, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | | | - Michael Müther
- Department of Neurosurgery, University Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| |
Collapse
|
39
|
Li K, Zheng Y, Cai S, Fan Z, Yang J, Liu Y, Liang S, Song M, Du S, Qi L. The subventricular zone structure, function and implications for neurological disease. Genes Dis 2025; 12:101398. [PMID: 39935607 PMCID: PMC11810716 DOI: 10.1016/j.gendis.2024.101398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 05/28/2024] [Accepted: 07/28/2024] [Indexed: 02/13/2025] Open
Abstract
The subventricular zone (SVZ) is a region surrounding the lateral ventricles that contains neural stem cells and neural progenitor cells, which can proliferate and differentiate into various neural and glial cells. SVZ cells play important roles in neurological diseases like neurodegeneration, neural injury, and glioblastoma multiforme. Investigating the anatomy, structure, composition, physiology, disease associations, and related mechanisms of SVZ is significant for neural stem cell therapy and treatment/prevention of neurological disorders. However, challenges remain regarding the mechanisms regulating SVZ cell proliferation, differentiation, and migration, delivering cells to damaged areas, and immune responses. In-depth studies of SVZ functions and related therapeutic developments may provide new insights and approaches for treating brain injuries and degenerative diseases, as well as a scientific basis for neural stem cell therapy. This review summarizes research findings on SVZ and neurological diseases to provide references for relevant therapies.
Collapse
Affiliation(s)
- Kaishu Li
- Department of Neurosurgery, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
- Institute of Digestive Diseases, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Yin Zheng
- Department of Neurosurgery, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
- Institute of Digestive Diseases, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Shubing Cai
- Department of Neurosurgery, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
- Institute of Digestive Diseases, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Zhiming Fan
- Department of Neurosurgery, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
- Institute of Digestive Diseases, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Junyi Yang
- Department of Neurosurgery, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
- Institute of Digestive Diseases, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Yuanrun Liu
- Department of Neurosurgery, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
- Institute of Digestive Diseases, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Shengqi Liang
- Department of Neurosurgery, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
- Institute of Digestive Diseases, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Meihui Song
- Department of Neurosurgery, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
- Institute of Digestive Diseases, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Siyuan Du
- Department of Neurosurgery, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
- Institute of Digestive Diseases, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| | - Ling Qi
- Institute of Digestive Diseases, The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, China
| |
Collapse
|
40
|
Moon HH, Wongsawaeng Wave D, Park JE, Park SY, Baek S, Kim YH, Song SW, Hong CK, Kim JH, Lee MH, Park YW, Ahn SS, Pollock JM, Barajas RF, Kim HS. Maximum Resection of Noncontrast-enhanced Tumor at MRI Is a Favorable Prognostic Factor in IDH Wild-Type Glioblastoma. Radiology 2025; 315:e241393. [PMID: 40326876 DOI: 10.1148/radiol.241393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Background Isocitrate dehydrogenase (IDH) wild-type glioblastoma often includes a noncontrast-enhanced tumor (NET) component, and the extent of NET resection may serve as a prognostic marker. Purpose To assess clinical outcomes based on gross total resection (GTR) of NET, develop a real-world survival model incorporating GTR-NET for IDH wild-type glioblastoma, and validate the findings in multinational external cohorts. Materials and Methods A retrospective analysis included patients with IDH wild-type glioblastoma in a prospective registry (March 2017 to October 2020) as the training set. External validation used consecutive patients from two centers (March 2017 to January 2023). Patients were stratified into three groups: GTR-NET, GTR in contrast-enhanced tumor (CET) only, and no GTR. A conditional inference tree (CIT) model was developed using GTR type, age, and O6-methylguanine DNA methyltransferase (MGMT) promoter methylation status to predict overall survival (OS) and was externally validated. Kaplan-Meier analysis, log-rank test, time-dependent area under the receiver operating characteristic curve, and Harrell C-indexes were used for evaluation. Results In the training set (n = 201; mean age, 60 years ± 11.3; 109 males), four survival groups were identified. GTR-NET was associated with longer OS (median, 32.6 months; IQR, 18.7-46.7 months; P < .001). When GTR-NET was not achieved, OS was stratified as follows: younger than age 60 years (median OS, 23.4 months; IQR, 12.2-34.8 months), age 60 years or older and positive for MGMT (median OS, 19.1 months; IQR, 13.0-27.8 months), and age 60 years or older and negative for MGMT (median OS, 10.7 months; IQR, 6.5-14.1 months). External validation sets (352 patients in external validation set 1 and 60 patients external validation set 2) confirmed these groups (P < .001 and P = .04). Time-dependent areas under the receiver operating characteristic curve ranged from 0.684 (95% CI: 0.623, 0.745) to 0.694 (95% CI: 0.631, 0.758) and from 0.610 (95% CI: 0.449, 0.771) to 0.678 (95% CI: 0.512, 0.844), with CIT sensitivity for GTR-NET at 70.7%-77.3% and 87.6%-87.9% and C-indexes of 0.65 and 0.63. Conclusion A GTR-NET-based survival model was developed and validated, demonstrating that GTR-NET is an independent prognostic marker for longer OS in IDH-wildtype glioblastoma. ClinicalTrials.gov identifier: NCT02619890 © RSNA, 2025 Supplemental material is available for this article.
Collapse
Affiliation(s)
- Hye Hyeon Moon
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea
| | | | - Ji Eun Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea
| | - Seo Young Park
- Department of Statistics and Data Science, Korea National Open University, Seoul, Korea
| | - Seunghee Baek
- Department of Statistics and Epidemiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Young-Hoon Kim
- Department of Neurosurgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Sang Woo Song
- Department of Neurosurgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Chang-Ki Hong
- Department of Neurosurgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Jeong Hoon Kim
- Department of Neurosurgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Myung Hwan Lee
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Yae Won Park
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung Soo Ahn
- Department of Radiology and Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yonsei University College of Medicine, Seoul, South Korea
| | | | - Ramon Francisco Barajas
- Department of Radiology, Oregon Health and Science University, Portland, Ore
- Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore
- Knight Cancer Institute, Oregon Health and Science University, Portland, Ore
| | - Ho Sung Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 43 Olympic-ro 88, Songpa-Gu, Seoul 05505, Korea
| |
Collapse
|
41
|
Kim YK, Song J. Metabolic imbalance and brain tumors: The interlinking metabolic pathways and therapeutic actions of antidiabetic drugs. Pharmacol Res 2025; 215:107719. [PMID: 40174814 DOI: 10.1016/j.phrs.2025.107719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025]
Abstract
Brain tumors are complex, heterogeneous malignancies, often associated with significant morbidity and mortality. Emerging evidence suggests the important role of metabolic syndrome, such as that observed in diabetes mellitus, in the progression of brain tumors. Several studies indicated that hyperglycemia, insulin resistance, oxidative stress, and altered adipokine profiles influence tumor growth, proliferation, and treatment resistance. Intriguingly, antidiabetic drugs (e.g., metformin, sulfonylureas, dipeptidyl peptidase-4 (DPP-4) inhibitors, glucagon-like peptide-1 (GLP-1) receptor agonists, and thiazolidinediones) have shown promise as adjunctive or repurposed agents in managing brain tumors. Metformin can impair tumor cell proliferation, enhance treatment sensitivity, and modify the tumor microenvironment by activating AMP-activated protein kinase (AMPK) and inhibiting mammalian target of rapamycin (mTOR) signaling pathways. DPP-4 inhibitors and GLP-1 receptor agonists can target both metabolic and inflammatory aspects of brain tumors, while thiazolidinediones may induce apoptosis in tumor cells and synergize with other therapeutics. Consequently, further studies and clinical trials are needed to confirm the efficacy, safety, and utility of metabolic interventions in treating brain tumors. Here, we review the evidence for the metabolic interconnections between metabolic diseases and brain tumors and multiple actions of anti-diabetes drugs in brain tumors.
Collapse
Affiliation(s)
- Young-Kook Kim
- Department of Biochemistry, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea.
| |
Collapse
|
42
|
Sun J, Zhao W, Zhang L, Wu S, Xue S, Cao H, Xu B, Li X, Hu N, Jiang T, Xu Y, Wang Z, Zhang C, Ren J. Centromere protein U mediates the ubiquitination and degradation of RPS3 to facilitate temozolomide resistance in glioblastoma. Drug Resist Updat 2025; 80:101214. [PMID: 40023134 DOI: 10.1016/j.drup.2025.101214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/10/2025] [Accepted: 02/15/2025] [Indexed: 03/04/2025]
Abstract
AIMS Temozolomide (TMZ) is the first-line chemotherapeutic agent for glioblastoma (GBM) therapy; however, resistance to TMZ remains a major obstacle in GBM treatment. The aim of this study is to elucidate the mechanisms underlying TMZ resistance and explore how to enhance the sensitivity of GBM to TMZ. METHODS GBM organoids were generated from patient samples, and organoid-based TMZ sensitivity testing was performed. Transcriptome sequencing was conducted on GBM organoids, which identified Centromere protein U (CENPU) as a novel key gene mediating TMZ resistance. Histopathological assessments were carried out using immunohistochemistry (IHC) and Hematoxylin and Eosin (HE) staining. Single-cell sequencing data were utilized to determine the functional states of CENPU in GBM cells. Intracranial and subcutaneous glioma mouse models were constructed to evaluate the effect of CENPU on TMZ sensitivity. The underlying mechanisms were further investigated using immunofluorescence, lentivirus transduction, co-immunoprecipitation, mass spectrometry, alkaline comet assay et al. RESULTS: CENPU was found to be highly expressed in TMZ-resistant GBM organoids and enhanced the TMZ resistance of GBM cells by promoting DNA damage repair. Its abnormal expression correlates with poor clinical outcomes in glioma patients. In vivo studies demonstrated that downregulation of CENPU enhances the sensitivity of GBM to TMZ. Correspondingly, rescue of CENPU expression reversed this effect on TMZ sensitivity in GBM cells. Mechanistically, CENPU cooperates with TRIM5α to promote the ubiquitination and degradation of RPS3 by inducing its polyubiquitination at the K214 residue. This process subsequently activates the ERK1/2 pathway and promotes the expression of E2F1 and RAD51. Consequently, the degradation of RPS3 and upregulation of RAD51 in GBM cells enhance DNA damage repair, thereby contributing to TMZ resistance. CONCLUSION Our study identified CENPU as a novel key gene mediating TMZ resistance and elucidated its molecular mechanisms, providing a new target to overcome TMZ resistance in GBM.
Collapse
Affiliation(s)
- Jinmin Sun
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China; Laboratory of Clinical and Experimental Pathology, Department of Pathology, Xuzhou Medical University, Xuzhou 221004, China
| | - Wenyu Zhao
- Laboratory of Clinical and Experimental Pathology, Department of Pathology, Xuzhou Medical University, Xuzhou 221004, China
| | - Lei Zhang
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Sicheng Wu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China
| | - Senrui Xue
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China
| | - Haowei Cao
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China
| | - Biao Xu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China
| | - Xinmiao Li
- Laboratory of Clinical and Experimental Pathology, Department of Pathology, Xuzhou Medical University, Xuzhou 221004, China
| | - Nan Hu
- Laboratory of Clinical and Experimental Pathology, Department of Pathology, Xuzhou Medical University, Xuzhou 221004, China
| | - Tao Jiang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Yixin Xu
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, China
| | - Zhifei Wang
- Department of Neurosurgery, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Chao Zhang
- Department of Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong, Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| | - Jing Ren
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou 221004, China.
| |
Collapse
|
43
|
Cao C, Wang L, Jiang F, Jin Q, Jin T, Huang S, Hu Q, Chen Y, Piao Y, Hua Y, Feng X, Zhou Y, Chen X. Granulocyte-macrophage colony-stimulating factor for newly diagnosed glioblastoma. Neoplasia 2025; 63:101156. [PMID: 40090128 PMCID: PMC11957598 DOI: 10.1016/j.neo.2025.101156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND There is a clear need to improve the efficiency of therapeutic strategy for patients with newly diagnosed glioblastoma (GBM). The purpose of this study was to evaluate the feasibility of hypofractionated intensity-modulated radiation therapy (IMRT), temozolomide and granulocyte-macrophage colony-stimulating factor (GM-CSF) for patients with newly diagnosed GBM. METHODS Patients were treated with hypofractionated IMRT (15 × 3.5Gy to the high-risk region and 15 × 3.0Gy to the low-risk region), temozolomide (75 mg per square meter of body-surface area per day, from 1 week before the beginning of radiotherapy to the last day of radiotherapy) and GM-CSF [200μg (equivalent to 125 μg/m² calculated dose) subcutaneously injected daily for 2 weeks, starting from the second week of radiotherapy]. The primary endpoint was 6-month progression free survival (PFS). RESULTS Between June 2016 and Feburary 2020, 41 patients were enrolled. During concomitant chemoradiotherapy, no grade 3 or 4 hematologic toxicities were observed and grade 3 non-hematologic toxicities were documented in 5 patients (12.2 %) due to GM-CSF. All patients completed both radiotherapy and concomitant temozolomide as planned. Only five patients (12.2 %) discontinued concomitant GM-CSF because of toxicity. At a median follow-up of 33.1 months (IQR 23.0-51.2), the 6-month PFS rate was 68.3 % (95 % CI: 54.0-82.6). The median overall survival of all patients was 16.7 months (95 % CI: 10.5-22.9). Compared with pre-GM-CSF, the concentrations of TNF-α (p = 1.9615E-10) and IL-18 (p = 6.8467E-8) were increased after GM-CSF, while the proportion of CD19 (p = 0.000015), the concentrations of IgG (p = 0.000015) and CXCL12 (p = 0.000257) were decreased. CONCLUSIONS The combination of hypofractionated IMRT, temozolomide and GM-CSF for GBM was feasible and safe. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02663440.
Collapse
Affiliation(s)
- Caineng Cao
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences; Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, China
| | - Le Wang
- Department of Cancer Prevention, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, China
| | - Feng Jiang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences; Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, China
| | - Qifeng Jin
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences; Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, China
| | - Ting Jin
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences; Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, China
| | - Shuang Huang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences; Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, China
| | - Qiaoying Hu
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences; Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, China
| | - Yuanyuan Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences; Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, China
| | - Yongfeng Piao
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences; Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, China
| | - Yonghong Hua
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences; Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, China
| | - Xinglai Feng
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences; Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, China
| | - Yi Zhou
- Department of Hematology & Oncology, Xiamen Amoytop Biotech Co., Ltd., China
| | - Xiaozhong Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences; Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, China.
| |
Collapse
|
44
|
Bisoi A, Majumdar T, Sarkar S, Singh PC. Nucleobase Level Information into the Folding of G-Quadruplex by Anti-inflammatory Drugs in the Absence of Salt. J Phys Chem B 2025; 129:4159-4168. [PMID: 40262105 DOI: 10.1021/acs.jpcb.5c00526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
G-quadruplexes (G4s) in the telomere region are important targets for cancer therapy. Molecules that can fold and stabilize the telomere DNA sequences, even in the absence of salt, can be an exciting prospect for therapy purposes. Anti-inflammatory drugs hydroxychloroquine (HCQ) and chloroquine (CQ) have shown promising effects in cancer therapy and also in the different levels of trial stages. In this study, we have investigated the structure and stability of several natural and mutated telomeric sequences with anti-inflammatory drugs and their analogues in the absence of salts using the biophysical and docking methods to understand the role of the quartet and loop nucleobases of DNA along with the functional group of drugs responsible for triggering the folding of telomeric DNA sequences into G4. The findings indicate that the hydrogen bonding between the charged side chain with the guanine repeating unit associated with the quartet and the thymine in the terminal loops of telomere DNA is the main driving force for the folding of telomere DNA sequences into G4 induced by anti-inflammatory drugs. The data indicate that the adenine nucleobase in the loop of the telomere does not play any role in its folding process induced by HCQ and CQ.
Collapse
Affiliation(s)
- Asim Bisoi
- School of the Chemical Science, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Trideep Majumdar
- School of the Chemical Science, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Sunipa Sarkar
- School of the Chemical Science, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Prashant Chandra Singh
- School of the Chemical Science, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| |
Collapse
|
45
|
Viozzi I, Hannink G, Ardon H, Balvers RK, Bosscher L, van Geest S, Ho VKY, Hovinga K, Kwee L, Tewarie RN, Robe PA, van der Veer O, Wagemakers M, Ter Laan M, De Witt Hamer PC. Between-hospital variation in biopsy indication for patients with newly diagnosed glioblastoma in the Dutch Quality Registry for Neurosurgery. J Neurooncol 2025; 172:625-632. [PMID: 39913047 PMCID: PMC11968504 DOI: 10.1007/s11060-025-04959-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 01/28/2025] [Indexed: 02/07/2025]
Abstract
PURPOSE This registry-based study aims to examine the variation in biopsy and resection indications for glioblastoma patients across Dutch hospitals and to identify patient- and hospital-related factors associated with the surgical treatment. METHODS Data from all 7443 adults with first-time glioblastoma surgery at 12 hospitals were obtained from the prospective population-based Quality Registry Neurosurgery in the Netherlands between 2011 and 2021. Patients were stratified by either biopsy or resection. We analyzed variation in American Association of Anesthesiologist (ASA) classification, Karnofsky Performance Score (KPS), gender and age distribution between the different centers. Between-hospital variation in biopsy percentage was analyzed using a funnel plot. Logistic regression was used to identify associated patient- and hospital-related factors. RESULTS In total, 32% of the newly diagnosed glioblastoma patients underwent a biopsy, with wide variations between the different centers (23-56%). Patients-related variables such as higher age or ASA classification and lower KPS were significantly associated with the indication for biopsy. After correction for these factors, between-hospital variation persisted, with two institutes performing more biopsies than expected and one less than expected. Median overall survival was 12.5 months (95% CI 12.2-12.9) in the resection group and 5.6 months (95% CI 5.1-6) in the biopsy group, with wide variations between the different centers. CONCLUSION A substantial between-hospital variation in biopsy percentages was found. Patient factors (age, ASA classification and KPS) but also hospital factors (such as academic setting) impact surgical decisions. Variation persisted also after correction for potential confounders, indicating that other factors play a role in decision-making.
Collapse
Affiliation(s)
- Ilaria Viozzi
- Department of Neurosurgery, Radboud University Medical Center, Radboud Institute for Health Sciences, 6525 GA, Nijmegen, The Netherlands.
| | - Gerjon Hannink
- Department of Medical Imaging, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Hilko Ardon
- Department of Neurosurgery, Elisabeth-Tweesteden Hospital, Tilburg, The Netherlands
| | - Rutger K Balvers
- Department of Neurosurgery, Brain Tumour Centre, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Lisette Bosscher
- Department of Neurosurgery Isala Hospital, Zwolle, The Netherlands
| | - Sarita van Geest
- Department of Neurosurgery, Martini Hospital, Groningen, The Netherlands
| | - Vincent K Y Ho
- Department of Research & Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
| | - Koos Hovinga
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Lesley Kwee
- Department of Neurosurgery, Northwest Clinics, Alkmaar, The Netherlands
| | - Rishi Nandoe Tewarie
- Department of Neurosurgery, Haaglanden Medical Centre, The Hague, The Netherlands
| | - Pierre A Robe
- Department of Neurosurgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | | | - Michiel Wagemakers
- Department of Neurosurgery, University Medical Centre Groningen, Groningen, The Netherlands
| | - Mark Ter Laan
- Department of Neurosurgery, Radboud University Medical Center, Radboud Institute for Health Sciences, 6525 GA, Nijmegen, The Netherlands
| | - Philip C De Witt Hamer
- Department of Neurosurgery, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| |
Collapse
|
46
|
Mousavi R, Soltani M, Souri M. Microneedle patch capable of dual drug release for drug delivery to brain tumors. Drug Deliv Transl Res 2025; 15:1567-1594. [PMID: 39186235 DOI: 10.1007/s13346-024-01696-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2024] [Indexed: 08/27/2024]
Abstract
Primary brain tumors are mostly managed using surgical resection procedures. Nevertheless, in certain cases, a thin layer of tumors may remain outside of the resection process due to the possibility of permanent injury; these residual tumors expose patients to the risk of tumor recurrence. This study has introduced the use of microneedle patches implanted after surgery with a dual-release mechanism for the administration of doxorubicin. The proposed patches possess the capability to administer drugs directly to the residual tumors and initiate chemotherapy immediately following surgical procedures. Three-dimensional simulation of drug delivery to residual tumors in the brain has been performed based on a finite element method. The impact of four important parameters on drug delivery has been investigated, involving the fraction of drug released in the burst phase, the density of microneedles on the patch, the length of microneedles, and the microvascular density of the tumor. The simulation findings indicate that lowering the fraction of drug released in the initial burst phase reduces the maximum average concentration, but the sustained release that continues for a longer period, increasing the bioavailability of free drug. However, the area under curve (AUC) for different release rates remains unchanged due to the fact that an identical dose of drug is supplied in each instance. By increasing the density of microneedles on the patch, concentration accumulation is provided over an extensive region of tumor, which in turn induces more cancer cell death. A comparative analysis of various lengths reveals that longer microneedles facilitate profound penetration into the tumor layers and present better therapeutic response due to extensive area of the tumor which is exposure to chemotherapeutic drugs. Furthermore, high microvascular density, as a characteristic of the tumor microenvironment, is shown to have a significant impact on the blood microvessels drainage of drugs and consequently lower therapeutic response outcome. Our approach offers a computational framework for creating localized drug delivery systems and addressing the challenges related to residual brain tumors.
Collapse
Affiliation(s)
- Robab Mousavi
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | - Madjid Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran.
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Canada.
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, Canada.
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada.
- Centre for Sustainable Business, International Business University, Toronto, Canada.
| | - Mohammad Souri
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| |
Collapse
|
47
|
Bhutani B, Sharma V, Ganguly NK, Rana R. Unravelling the modified T cell receptor through Gen-Next CAR T cell therapy in Glioblastoma: Current status and future challenges. Biomed Pharmacother 2025; 186:117987. [PMID: 40117901 DOI: 10.1016/j.biopha.2025.117987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025] Open
Abstract
PURPOSE Despite current technological advancements in the treatment of glioma, immediate alleviation of symptoms can be catered by therapeutic modalities, including surgery, chemotherapy, and combinatorial radiotherapy that exploit aberrations of glioma. Additionally, a small number of target antigens, their heterogeneity, and immune evasion are the potential reasons for developing targeted therapies. This oncologic milestone has catalyzed interest in developing immunotherapies against Glioblastoma to improve overall survival and cure patients with high-grade glioma. The next-gen CAR-T Cell therapy is one of the effective immunotherapeutic strategies in which autologous T cells have been modified to express receptors against GBM and it modulates cytotoxicity. METHODS In this review article, we examine preclinical and clinical outcomes, and limitations as well as present cutting-edge techniques to improve the function of CAR-T cell therapy and explore the possibility of combination therapy. FINDINGS To date, several CAR T-cell therapies are being evaluated in clinical trials for GBM and other brain malignancies and multiple preclinical studies have demonstrated encouraging outcomes. IMPLICATIONS CAR-T cell therapy represents a promising therapeutic paradigm in the treatment of solid tumors but a few limitations include, the blood-brain barrier (BBB), antigen escape, tumor microenvironment (TME), tumor heterogeneity, and its plasticity that suppresses immune responses weakens the ability of this therapy. Additional investigation is required that can accurately identify the targets and reflect the similar architecture of glioblastoma, thus optimizing the efficiency of CAR-T cell therapy; allowing for the selection of patients most likely to benefit from immuno-based treatments.
Collapse
Affiliation(s)
- Bhavya Bhutani
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Vyoma Sharma
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Nirmal Kumar Ganguly
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Rashmi Rana
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India.
| |
Collapse
|
48
|
Kapteijn MY, Bakker N, Koekkoek JAF, Versteeg HH, Buijs JT. Venous Thromboembolism in Patients with Glioblastoma: Molecular Mechanisms and Clinical Implications. Thromb Haemost 2025; 125:421-434. [PMID: 39168144 PMCID: PMC12040436 DOI: 10.1055/s-0044-1789592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 08/06/2024] [Indexed: 08/23/2024]
Abstract
Patients with glioblastoma are among the cancer patients with the highest risk of developing venous thromboembolism (VTE). Long-term thromboprophylaxis is not generally prescribed because of the increased susceptibility of glioblastoma patients to intracranial hemorrhage. This review provides an overview of the current clinical standard for glioblastoma patients, as well as the molecular and genetic background which underlies the high incidence of VTE. The two main procoagulant proteins involved in glioblastoma-related VTE, podoplanin and tissue factor, are described, in addition to the genetic aberrations that can be linked to a hypercoagulable state in glioblastoma. Furthermore, possible novel biomarkers and future treatment strategies are discussed, along with the potential of sequencing approaches toward personalized risk prediction for VTE. A glioblastoma-specific VTE risk stratification model may help identifying those patients in which the increased risk of bleeding due to extended anticoagulation is outweighed by the decreased risk of VTE.
Collapse
Affiliation(s)
- Maaike Y. Kapteijn
- Division of Thrombosis and Hemostasis, Department of Medicine, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Nina Bakker
- Division of Thrombosis and Hemostasis, Department of Medicine, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Johan A. F. Koekkoek
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Henri H. Versteeg
- Division of Thrombosis and Hemostasis, Department of Medicine, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeroen T. Buijs
- Division of Thrombosis and Hemostasis, Department of Medicine, Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
49
|
Chithra P, Bhatia D, Solanki R. Advanced nanomicelles for targeted glioblastoma multiforme therapy. BIOMATERIALS ADVANCES 2025; 170:214221. [PMID: 39922136 DOI: 10.1016/j.bioadv.2025.214221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/11/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive and malignant primary brain tumor, classified as grade IV by the WHO. Despite standard treatments like surgical resection, radiotherapy and chemotherapy (i.e. temozolomide), GBM's prognosis remains poor due to its heterogeneity, recurrence and the impermeability of the blood-brain barrier (BBB). The exact cause of GBM is unclear with potential factors including genetic predisposition and ionizing radiation. Innovative approaches such as nanomicelles-nanoscale, self-assembled structures made from lipids and amphiphilic polymers show promise for GBM therapy. These nanocarriers enhance drug solubility and stability, enabling targeted delivery of therapeutic agents across the BBB. This review explores the synthesis strategies, characterization and applications of nanomicelles in GBM treatment. Nanomicelles improve the delivery of both hydrophobic and hydrophilic drugs and provide non-invasive delivery options. By offering site-specific targeting, biocompatibility, and stability, nanomicelles can potentially overcome the limitations of current GBM therapies. This review highlights recent advancements in the use of nanomicelles for delivering therapeutic agents and nucleic acids addressing the critical need for advanced treatments to improve GBM patient outcomes.
Collapse
Affiliation(s)
- P Chithra
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Raghu Solanki
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| |
Collapse
|
50
|
Pham TN, Coupey J, Yger F, Candéias SM, Thariat J, Valable S. Effect of glioblastoma and brain radiotherapy on T-lymphocyte subpopulations in rodents. Radiother Oncol 2025; 206:110801. [PMID: 40081500 DOI: 10.1016/j.radonc.2025.110801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/07/2025] [Accepted: 02/20/2025] [Indexed: 03/16/2025]
Abstract
INTRODUCTION Although lymphopenia is linked to immune suppression favoring tumor growth, the effect of different radiation types on specific T-lymphocyte subsets remains unclear. Among the T-lymphocyte subpopulations, CD8+-lymphocytes serve as key effectors in cancer immunity. This study aimed to examine the changes in T-lymphocyte subpopulations in both tumor-free and glioblastoma-bearing mice following brain-irradiation. METHODS The study was divided into two main phases. First, C57BL/6 mice, with or without glioblastoma (GL261 cells), received hemispheric brain-irradiation or no-treatment. T-lymphocyte subpopulations were analyzed using flow cytometry at various timepoint. The effect of tumor size and brain-irradiation on these cells was assessed using correlation analysis. Next, C57BL/6 mice were subjected to different brain-irradiation conditions. Blood samples were collected during and post-irradiation to analyze T-lymphocyte subpopulations, and tree-based modeling was used to determine radiation parameters impact on naïve CD8+-lymphocyte levels. RESULTS Glioblastoma reduced all T-lymphocyte subpopulations by day 15 post-inoculation. Radiotherapy decreased regulatory and effector CD4+-lymphocytes in both tumor-free and glioblastoma-bearing mice, but not naïve or memory CD4+-lymphocytes, in both tumor-free and glioblastoma-bearing mice. In tumor-free mice, radiotherapy had no effect on CD8+-lymphocytes, but reduced all CD8+-lymphocyte types in glioblastoma-bearing mice. Glioblastoma size negatively affected CD8+-lymphocytes. Brain-irradiation persistently reduced naïve and memory CD8+-lymphocytes, but effector and regulatory T-lymphocytes recovered. Exposure of lymph nodes to radiation worsened CD8+-lymphocyte reduction. CONCLUSION These findings confirm that the presence of glioblastoma and brain-irradiation affect T-lymphocyte subpopulations in mice. The inclusion of lymph nodes in the irradiated area led to a long-term decrease in naïve CD8+-lymphocytes. Further mechanistic studies are needed to understand the molecular basis of radiation impact on T-lymphocyte subpopulations.
Collapse
Affiliation(s)
- Thao-Nguyen Pham
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, F-14000 Caen, France; Laboratoire de Physique Corpusculaire UMR6534 IN2P3/ENSICAEN, France - Normandie Université, France
| | - Julie Coupey
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, F-14000 Caen, France
| | - Florian Yger
- Univ. Paris-Dauphine, PSL Research Univ./CNRS, LAMSADE, Paris, France
| | - Serge M Candéias
- Univ. Grenoble Alpes, CEA, CNRS, IRIG-LCBM-UMR5249, 38054 Grenoble, France
| | - Juliette Thariat
- Laboratoire de Physique Corpusculaire UMR6534 IN2P3/ENSICAEN, France - Normandie Université, France; Department of Radiation Oncology, Centre François Baclesse, Caen, Normandy, France.
| | - Samuel Valable
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, F-14000 Caen, France.
| |
Collapse
|