1
|
Janowicz PW, Boele T, Maschmeyer RT, Gholami YH, Kempe EG, Stringer BW, Stoner SP, Zhang M, du Toit-Thompson T, Williams F, Touffu A, Munoz L, Kuncic Z, Brighi C, Waddington DEJ. Enhanced detection of glioblastoma vasculature with superparamagnetic iron oxide nanoparticles and MRI. Sci Rep 2025; 15:14283. [PMID: 40274951 PMCID: PMC12022243 DOI: 10.1038/s41598-025-97943-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: 01/22/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
Detecting glioblastoma infiltration in the brain is challenging due to limited MRI contrast beyond the enhancing tumour core. This study aims to investigate the potential of superparamagnetic iron oxide nanoparticles (SPIONs) as contrast agents for improved detection of diffuse brain cancer. We examine the distribution and pharmacokinetics of SPIONs in glioblastoma models with intact and disrupted blood-brain barriers. Using MRI, we imaged RN1-luc and U87MG mice injected with Gadovist and SPIONs, observing differences in blood-brain barrier permeability. Peripheral imaging showed strong uptake of nanoparticles in the liver and spleen, while vascular and renal signals were transient. Susceptibility gradient mapping enabled positive nanoparticle contrast within tumours and provided additional information on tumour angiogenesis. This approach offers a novel method for detecting diffuse brain cancer. Our findings demonstrate that SPIONs enhance glioblastoma detection beyond conventional MRI, providing insights into tumour angiogenesis and opening new avenues for early diagnosis and targeted treatment strategies.
Collapse
Affiliation(s)
- Phillip W Janowicz
- Image X Institute, Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.
| | - Thomas Boele
- Image X Institute, Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Richard T Maschmeyer
- School of Physics, Faculty of Science, The University of Sydney, Sydney, Australia
| | - Yaser H Gholami
- School of Physics, Faculty of Science, The University of Sydney, Sydney, Australia
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, Australia
| | - Emma G Kempe
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Brett W Stringer
- Institute for Biomedicine and Glycomics, Griffith University, Brisbane, Australia
| | - Shihani P Stoner
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, The University of Sydney, Royal North Shore Hospital, Sydney, Australia
| | - Marie Zhang
- Imagion Biosystems Ltd, Melbourne, Australia
| | - Taymin du Toit-Thompson
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, The University of Sydney, Royal North Shore Hospital, Sydney, Australia
| | - Fern Williams
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, The University of Sydney, Royal North Shore Hospital, Sydney, Australia
| | - Aude Touffu
- Bill Walsh Translational Cancer Research Laboratory, Kolling Institute of Medical Research, The University of Sydney, Royal North Shore Hospital, Sydney, Australia
| | - Lenka Munoz
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Zdenka Kuncic
- School of Physics, Faculty of Science, The University of Sydney, Sydney, Australia
- The Sydney Nano Institute, The University of Sydney, Sydney, Australia
| | - Caterina Brighi
- Image X Institute, Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - David E J Waddington
- Image X Institute, Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.
| |
Collapse
|
2
|
Song X, Tiek D, Lu M, Yu X, Wu R, Walker M, He Q, Sisbarro D, Hu B, Cheng SY. A Single-Cell Atlas of RNA Alternative Splicing in the Glioma-Immune Ecosystem. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645511. [PMID: 40196477 PMCID: PMC11974875 DOI: 10.1101/2025.03.26.645511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Single-cell analysis has refined our understanding of cellular heterogeneity in glioma, yet RNA alternative splicing (AS)-a critical layer of transcriptome regulation-remains underexplored at single-cell resolution. Here, we present a pan-glioma single-cell AS analysis in both tumor and immune cells through integrating seven SMART-seq2 datasets of human gliomas. Our analysis reveals lineage-specific AS across glioma cellular states, with the most divergent AS landscapes between mesenchymal- and neuronal-like glioma cells, exemplified by AS in TCF12 and PTBP2. Comparison between core and peripheral glioma cells highlights AS-redox co-regulation of cytoskeleton organization. Further analysis of glioma-infiltrating immune cells reveals potential isoform-level regulation of protein glycosylation in regulatory T cells and a link between MS4A7 AS in macrophages and clinical response to anti-PD-1 therapy. This study emphasizes the role of AS in glioma cellular heterogeneity, highlighting the importance of an isoform-centric approach to better understand the complex biological processes driving tumorigenesis.
Collapse
Affiliation(s)
- Xiao Song
- The Ken & Ruth Davee Department of Neurology, The Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Deanna Tiek
- The Ken & Ruth Davee Department of Neurology, The Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Minghui Lu
- The Ken & Ruth Davee Department of Neurology, The Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Xiaozhou Yu
- The Ken & Ruth Davee Department of Neurology, The Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Runxin Wu
- The Ken & Ruth Davee Department of Neurology, The Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Maya Walker
- The Ken & Ruth Davee Department of Neurology, The Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Qiu He
- The Ken & Ruth Davee Department of Neurology, The Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Derek Sisbarro
- The Ken & Ruth Davee Department of Neurology, The Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Bo Hu
- The Ken & Ruth Davee Department of Neurology, The Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Shi-Yuan Cheng
- The Ken & Ruth Davee Department of Neurology, The Lou and Jean Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
3
|
Hendriks TF, Birmpili A, de Vleeschouwer S, Heeren RM, Cuypers E. Integrating Rapid Evaporative Ionization Mass Spectrometry Classification with Matrix-Assisted Laser Desorption Ionization Mass Spectrometry Imaging and Liquid Chromatography-Tandem Mass Spectrometry to Unveil Glioblastoma Overall Survival Prediction. ACS Chem Neurosci 2025; 16:1021-1033. [PMID: 40007067 PMCID: PMC11926789 DOI: 10.1021/acschemneuro.4c00463] [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: 07/23/2024] [Revised: 01/24/2025] [Accepted: 02/19/2025] [Indexed: 02/27/2025] Open
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive brain cancer with a median survival of 15 months. Despite advancements in conventional treatment approaches such as surgery and chemotherapy, the prognosis remains poor. This study investigates the use of rapid evaporative ionization mass spectrometry (REIMS) for real-time overall survival time classification of GBM samples and uses matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI-MSI) to compare lipidomic differences within GBM tumors. A total of 45 GBM biopsies were analyzed to develop a survival prediction model for IDH-wild type GBM. REIMS patterns from 28 patients were classified with a 97.7% correct classification rate, identifying key discriminators between short-term (0-12 months) and prolonged (>12 months) survivors. Cross-validation with additional samples showed that the model correctly classified short-term and prolonged survival with 66.7 and 69.4% accuracy, respectively. MALDI-MSI was performed to confirm the discriminators derived from REIMS data. Results indicated 42 and 33 discriminating features for short-term and prolonged survival, respectively. Proteomic profiling was performed by isolating tumor regions via laser-capture microdissection (LMD) and liquid chromatography-tandem mass spectrometry (LC-MS/MS). Subsequently, 1387 proteins were identified, of which 79 were significantly altered. In conclusion, this study shows that REIMS rapidly predicts glioblastoma survival times based on lipidomic profiles during electrosurgical dissection. MALDI-MSI confirmed that these differences were specific to the tumor region in the glioblastoma sections. LMD-guided LC-MS/MS-based proteomics revealed significantly altered pathways between short-term and prolonged survival. This research, including the comprehensive predictive survival model for GBM, could guide tumor resection surgeries based on accurate real-time tumor tissue identification as well as provide insights into overall survival mechanisms, possibly related to therapy response.
Collapse
Affiliation(s)
- Tim F.E. Hendriks
- The Maastricht
MultiModal Molecular Imaging (M4I) Institute, Division of Imaging
Mass Spectrometry (IMS), Maastricht University, Maastricht 6229 ER, The Netherlands
| | - Angeliki Birmpili
- The Maastricht
MultiModal Molecular Imaging (M4I) Institute, Division of Imaging
Mass Spectrometry (IMS), Maastricht University, Maastricht 6229 ER, The Netherlands
| | - Steven de Vleeschouwer
- Department
of Neurosurgery, UZ Leuven, and Laboratory for Experimental Neurosurgery
and Neuroanatomy, Department of Neurosciences and Leuven Brain Institute
(LBI), KU Leuven, Leuven 3000, Belgium
| | - Ron M.A. Heeren
- The Maastricht
MultiModal Molecular Imaging (M4I) Institute, Division of Imaging
Mass Spectrometry (IMS), Maastricht University, Maastricht 6229 ER, The Netherlands
| | - Eva Cuypers
- The Maastricht
MultiModal Molecular Imaging (M4I) Institute, Division of Imaging
Mass Spectrometry (IMS), Maastricht University, Maastricht 6229 ER, The Netherlands
| |
Collapse
|
4
|
Fathi Kazerooni A, Akbari H, Hu X, Bommineni V, Grigoriadis D, Toorens E, Sako C, Mamourian E, Ballinger D, Sussman R, Singh A, Verginadis II, Dahmane N, Koumenis C, Binder ZA, Bagley SJ, Mohan S, Hatzigeorgiou A, O'Rourke DM, Ganguly T, De S, Bakas S, Nasrallah MP, Davatzikos C. The radiogenomic and spatiogenomic landscapes of glioblastoma and their relationship to oncogenic drivers. COMMUNICATIONS MEDICINE 2025; 5:55. [PMID: 40025245 PMCID: PMC11873127 DOI: 10.1038/s43856-025-00767-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 02/12/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Glioblastoma is a highly heterogeneous brain tumor, posing challenges for precision therapies and patient stratification in clinical trials. Understanding how genetic mutations influence tumor imaging may improve patient management and treatment outcomes. This study investigates the relationship between imaging features, spatial patterns of tumor location, and genetic alterations in IDH-wildtype glioblastoma, as well as the likely sequence of mutational events. METHODS We conducted a retrospective analysis of 357 IDH-wildtype glioblastomas with pre-operative multiparametric MRI and targeted genetic sequencing data. Radiogenomic signatures and spatial distribution maps were generated for key mutations in genes such as EGFR, PTEN, TP53, and NF1 and their corresponding pathways. Machine and deep learning models were used to identify imaging biomarkers and stratify tumors based on their genetic profiles and molecular heterogeneity. RESULTS Here, we show that glioblastoma mutations produce distinctive imaging signatures, which are more pronounced in tumors with less molecular heterogeneity. These signatures provide insights into how mutations affect tumor characteristics such as neovascularization, cell density, invasion, and vascular leakage. We also found that tumor location and spatial distribution correlate with genetic profiles, revealing associations between tumor regions and specific oncogenic drivers. Additionally, imaging features reflect the cross-sectionally inferred evolutionary trajectories of glioblastomas. CONCLUSIONS This study establishes clinically accessible imaging biomarkers that capture the molecular composition and oncogenic drivers of glioblastoma. These findings have potential implications for noninvasive tumor profiling, personalized therapies, and improved patient stratification in clinical trials.
Collapse
Affiliation(s)
- Anahita Fathi Kazerooni
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA
- Center for Data-Driven Discovery in Biomedicine (D3b), Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hamed Akbari
- Department of Bioengineering, School of Engineering, Santa Clara University, Santa Clara, CA, USA
| | - Xiaoju Hu
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ, USA
| | - Vikas Bommineni
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA
| | - Dimitris Grigoriadis
- Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
| | - Erik Toorens
- Penn Genomic Analysis Core, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chiharu Sako
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Mamourian
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dominique Ballinger
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robyn Sussman
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ashish Singh
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ioannis I Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nadia Dahmane
- Department of Neurological Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zev A Binder
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen J Bagley
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Suyash Mohan
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Artemis Hatzigeorgiou
- Department of Computer Science and Biomedical Informatics, University of Thessaly, Lamia, Greece
- Hellenic Pasteur Institute, Athens, Greece
| | - Donald M O'Rourke
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Tapan Ganguly
- Penn Genomic Analysis Core, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Subhajyoti De
- Rutgers Cancer Institute of New Jersey, Rutgers the State University of New Jersey, New Brunswick, NJ, USA
| | - Spyridon Bakas
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - MacLean P Nasrallah
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology & Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- AI2D Center for AI and Data Science for Integrated Diagnostics, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Sun J, Liang S, Liu X, Zhang S, Li M, Zhang Q, Chen J. Insights into the selectivity of a brain-penetrant CDK4/6 vs CDK1/2 inhibitor for glioblastoma used in multiple replica molecular dynamics simulations. J Biomol Struct Dyn 2025; 43:2223-2242. [PMID: 38112295 DOI: 10.1080/07391102.2023.2294175] [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: 07/27/2023] [Accepted: 11/23/2023] [Indexed: 12/21/2023]
Abstract
Cyclin dependent kinases (CDKs) play an important role in cell cycle regulation and their dysfunction is associated with many cancers. That is why CDKs have been attractive targets for the treatment of cancer. Glioblastoma is a cancer caused by the aberrant expression of CDK4/6, so exploring the mechanism of the selection of CDK4/6 toward inhibitors relative to the other family members CDK1/2 is essential. In this work, multiple replica molecular dynamics (MRMD) simulations, principal component analysis (PCA), free energy landscapes (FELs), molecular mechanics Poisson-Boltzmann/Generalized Born surface area (MM-PB/GBSA) and other methods were integrated to decipher the selectively binding mechanism of the inhibitor N1J to CDK4/6 and CDK1/2. Molecular electrostatic potential (MESP) analysis provides an explanation for the N1J selectivity. Residue-based free energy decomposition reveals that most of the hot residues are located at the same location of CDKs proteins, but the different types of residues in different proteins cause changes in binding energy, which is considered as a potential developmental direction to improve the selectivity of inhibitors to CDK4/6. These results provide insights into the source of inhibitor and CDK4/6 selectivity for the future development of more selective inhibitors.
Collapse
Affiliation(s)
- Jiahao Sun
- School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Shanshan Liang
- School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Xinguo Liu
- School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Shaolong Zhang
- School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Meng Li
- School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Qinggang Zhang
- School of Physics and Electronics, Shandong Normal University, Jinan, China
| | - Jianzhong Chen
- School of Science, Shandong Jiaotong University, Jinan, China
| |
Collapse
|
6
|
Kim J, Kim R, Lee W, Kim GH, Jeon S, Lee YJ, Lee JS, Kim KH, Won J, Lee W, Park K, Kim HJ, Im S, Lee KJ, Park C, Kim J, Lee JY. Assembly of glioblastoma tumoroids and cerebral organoids: a 3D in vitro model for tumor cell invasion. Mol Oncol 2025; 19:698-715. [PMID: 39473365 PMCID: PMC11887666 DOI: 10.1002/1878-0261.13740] [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: 04/09/2024] [Revised: 08/02/2024] [Accepted: 09/13/2024] [Indexed: 03/08/2025] Open
Abstract
Glioblastoma (GBM) has a fatal prognosis because of its aggressive and invasive characteristics. Understanding the mechanism of invasion necessitates an elucidation of the relationship between tumor cells and the tumor microenvironment. However, there has been a scarcity of suitable models to investigate this. In this study, we established a glioblastoma-cerebral organoid assembloid (GCOA) model by co-culturing patient-derived GBM tumoroids and human cerebral organoids. Tumor cells from the tumoroids infiltrated the cerebral organoids, mimicking the invasive nature of the parental tumors. Using time-lapse imaging, various invasion patterns of cancer cells within cerebral organoids resembling a normal tissue milieu were monitored. Both single- and collective-cell invasion was captured in real-time. We also confirmed the formation of an intercellular tumor network and tumor-normal-cell interactions. Furthermore, the transcriptomic characterization of GCOAs revealed distinct features of invasive tumor cells. Overall, this study established the GCOA as a three-dimensional (3D) in vitro assembloid model to investigate invasion mechanisms and interactions between tumor cells and their microenvironment.
Collapse
Affiliation(s)
- Jieun Kim
- Department of Anatomy and Cell BiologySeoul National University College of MedicineSeoulKorea
| | - Rokhyun Kim
- Medical Research CenterGenomic Medicine Institute, Seoul National UniversitySeoulKorea
- Department of Biomedical SciencesSeoul National University College of MedicineSeoulKorea
| | - Wonseok Lee
- Department of Anatomy and Cell BiologySeoul National University College of MedicineSeoulKorea
- Department of Transitional MedicineSeoul National University College of MedicineSeoulKorea
- Department of Neurosurgery, Seoul National University HospitalSeoul National University College of MedicineSeoulKorea
| | - Gyu Hyun Kim
- Laboratory of Synaptic Circuit Plasticity, Neural Circuits Research GroupKorea Brain Research InstituteDaeguKorea
| | - Seeun Jeon
- Department of Anatomy and Cell BiologySeoul National University College of MedicineSeoulKorea
| | - Yun Jin Lee
- Department of Anatomy and Cell BiologySeoul National University College of MedicineSeoulKorea
| | - Jong Seok Lee
- Division of Pediatric NeurosurgerySeoul National University Children's HospitalSeoulKorea
| | - Kyung Hyun Kim
- Department of Anatomy and Cell BiologySeoul National University College of MedicineSeoulKorea
- Division of Pediatric NeurosurgerySeoul National University Children's HospitalSeoulKorea
| | - Jae‐Kyung Won
- Department of Pathology, Seoul National University HospitalSeoul National University College of MedicineSeoulKorea
| | - Woochan Lee
- Medical Research CenterGenomic Medicine Institute, Seoul National UniversitySeoulKorea
- Department of Biomedical SciencesSeoul National University College of MedicineSeoulKorea
| | - Kyunghyuk Park
- Medical Research CenterGenomic Medicine Institute, Seoul National UniversitySeoulKorea
| | - Hyun Je Kim
- Department of Biomedical SciencesSeoul National University College of MedicineSeoulKorea
- Cancer Research Institute, Medical Research CenterSeoul National University College of MedicineSeoulKorea
| | - Sun‐Wha Im
- Department of Biochemistry and Molecular BiologyKangwon National University School of MedicineChuncheonKorea
| | - Kea Joo Lee
- Laboratory of Synaptic Circuit Plasticity, Neural Circuits Research GroupKorea Brain Research InstituteDaeguKorea
| | - Chul‐Kee Park
- Department of Neurosurgery, Seoul National University HospitalSeoul National University College of MedicineSeoulKorea
| | - Jong‐Il Kim
- Medical Research CenterGenomic Medicine Institute, Seoul National UniversitySeoulKorea
- Department of Biomedical SciencesSeoul National University College of MedicineSeoulKorea
- Cancer Research Institute, Medical Research CenterSeoul National University College of MedicineSeoulKorea
- Department of Biochemistry and Molecular BiologySeoul National University College of MedicineSeoulKorea
| | - Ji Yeoun Lee
- Department of Anatomy and Cell BiologySeoul National University College of MedicineSeoulKorea
- Division of Pediatric NeurosurgerySeoul National University Children's HospitalSeoulKorea
- Neuroscience Research Institute, Medical Research CenterSeoul National University College of MedicineSeoulKorea
| |
Collapse
|
7
|
Haydo A, Schmidt J, Crider A, Kögler T, Ertl J, Hehlgans S, Hoffmann ME, Rathore R, Güllülü Ö, Wang Y, Zhang X, Herold-Mende C, Pampaloni F, Tegeder I, Dikic I, Dai M, Rödel F, Kögel D, Linder B. BRAT1 - a new therapeutic target for glioblastoma. Cell Mol Life Sci 2025; 82:52. [PMID: 39833546 PMCID: PMC11747058 DOI: 10.1007/s00018-024-05553-0] [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: 07/15/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025]
Abstract
Glioblastoma (GBM), the most malignant primary brain tumor in adults, has poor prognosis irrespective of therapeutic advances due to its radio-resistance and infiltrative growth into brain tissue. The present study assessed functions and putative druggability of BRCA1-associated ATM activator 1 (BRAT1) as a crucial factor driving key aspects of GBM, including enhanced DNA damage response and tumor migration. By a stable depletion of BRAT1 in GBM and glioma stem-like (GSC) cell lines, we observed a delay in DNA double-strand break repair and increased sensitivity to radiation treatment, corroborated by in vitro and in vivo studies demonstrating impaired tumor growth and invasion. Proteomic and phosphoproteomic analyses further emphasize the role of BRAT1's cell migration and invasion capacity, with a notable proportion of downregulated proteins associated with these processes. In line with the genetic manipulation, we found that treatment with the BRAT1 inhibitor Curcusone D (CurD) significantly reduced GSC migration and invasion in an ex vivo slice culture model, particularly when combined with irradiation, resulting in a synergistic inhibition of tumor growth and infiltration. Our results reveal that BRAT1 contributes to GBM growth and invasion and suggest that therapeutic inhibition of BRAT1 with CurD or similar compounds might constitute a novel approach for anti-GBM directed treatments.
Collapse
Affiliation(s)
- Alicia Haydo
- Experimental Neurosurgery, Department of Neurosurgery, Neuroscience Center, Goethe University Hospital, Goethe University Frankfurt, 60528, Frankfurt am Main, Germany.
| | - Jennifer Schmidt
- Experimental Neurosurgery, Department of Neurosurgery, Neuroscience Center, Goethe University Hospital, Goethe University Frankfurt, 60528, Frankfurt am Main, Germany
| | - Alisha Crider
- Experimental Neurosurgery, Department of Neurosurgery, Neuroscience Center, Goethe University Hospital, Goethe University Frankfurt, 60528, Frankfurt am Main, Germany
| | - Tim Kögler
- Experimental Neurosurgery, Department of Neurosurgery, Neuroscience Center, Goethe University Hospital, Goethe University Frankfurt, 60528, Frankfurt am Main, Germany
| | - Johanna Ertl
- Experimental Neurosurgery, Department of Neurosurgery, Neuroscience Center, Goethe University Hospital, Goethe University Frankfurt, 60528, Frankfurt am Main, Germany
- Radiation Biology and DNA Repair, Darmstadt, TU, Germany
| | - Stephanie Hehlgans
- Department of Radiotherapy and Oncology, Goethe University Hospital, 60590, Frankfurt am Main, Germany
| | - Marina E Hoffmann
- Institute of Biochemistry II, Goethe University, Frankfurt am Main, Germany
| | - Rajeshwari Rathore
- Institute of Biochemistry II, Goethe University, Frankfurt am Main, Germany
| | - Ömer Güllülü
- Department of Radiotherapy and Oncology, Goethe University Hospital, 60590, Frankfurt am Main, Germany
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, USA
| | - Yecheng Wang
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, USA
| | - Xiangke Zhang
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, USA
| | - Christel Herold-Mende
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Francesco Pampaloni
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, 60438, Frankfurt am Main, Germany
| | - Irmgard Tegeder
- Institute for Clinical Pharmacology, Faculty of Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Ivan Dikic
- Institute of Biochemistry II, Goethe University, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, 60438, Frankfurt am Main, Germany
- Cardio-Pulmonary Institute, 60590, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, 60590, Frankfurt am Main, Germany
| | - Mingji Dai
- Department of Chemistry and Winship Cancer Institute, Emory University, Atlanta, GA, 30022, USA
| | - Franz Rödel
- Department of Radiotherapy and Oncology, Goethe University Hospital, 60590, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, 60590, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60590, Frankfurt am Main, Germany
- German Cancer Research Center DKFZ, 69120, Heidelberg, Germany
| | - Donat Kögel
- Experimental Neurosurgery, Department of Neurosurgery, Neuroscience Center, Goethe University Hospital, Goethe University Frankfurt, 60528, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60590, Frankfurt am Main, Germany
- German Cancer Research Center DKFZ, 69120, Heidelberg, Germany
| | - Benedikt Linder
- Experimental Neurosurgery, Department of Neurosurgery, Neuroscience Center, Goethe University Hospital, Goethe University Frankfurt, 60528, Frankfurt am Main, Germany
| |
Collapse
|
8
|
Li Y, Wang R, Chen J, Zhu Z, Wang Y, Ma W. 68Ga-NOTA-RM26 PET/CT in the evaluation of glioma: a pilot prospective study. EJNMMI Res 2025; 15:6. [PMID: 39821814 PMCID: PMC11748694 DOI: 10.1186/s13550-025-01198-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND Gliomas are the most common malignant primary tumors of the central nervous system. There is an urgent need for new convenient, targeted and specific imaging agents for gliomas. This study aimed to firstly evaluate the feasibility of 68Ga-NOTA-RM26 PET/CT imaging in glioma and analyze the relationship between the imaging characteristics and glioma grade, classification and molecular alterations. RESULTS Twenty-two patients were confirmed as glioma by surgery or biopsy. All patients exhibited 68Ga-NOTA-RM26 uptake. SUVmax was chosen as the imaging marker for analysis. For all glioma patients, there were significant differences between grades (P = 0.047). For primary gliomas, SUVmax had good discrimination for both tumor classifications (P = 0.045) and grades (P = 0.03). There was a positive correlation (P < 0.01) between GRPR expression level and SUVmax. P53 mutations caused significant differences in SUVmax (P = 0.03). CONCLUSIONS This study is the first application of 68Ga-NOTA-RM26 in glioma patients and confirmed the safety and efficacy in glioma patients. 68Ga-NOTA-RM26 PET/CT has potential value in tumor grade, classification, and molecular alterations. TRIAL REGISTRATION ClinicalTrials.gov: NCT06412952. Registered 26 April 2024, https://clinicaltrials.gov/study/NCT06412952.
Collapse
Affiliation(s)
- Yilin Li
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Rongxi Wang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jingci Chen
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhaohui Zhu
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Yu Wang
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Wenbin Ma
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT Alliance, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
9
|
Shojaee P, Weinholtz E, Schaadt NS, Feuerhake F, Hatzikirou H. Biopsy location and tumor-associated macrophages in predicting malignant glioma recurrence using an in-silico model. NPJ Syst Biol Appl 2025; 11:3. [PMID: 39779740 PMCID: PMC11711667 DOI: 10.1038/s41540-024-00478-7] [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: 06/22/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
Predicting the biological behavior and time to recurrence (TTR) of high-grade diffuse gliomas (HGG) after maximum safe neurosurgical resection and combined radiation and chemotherapy plays a pivotal role in planning clinical follow-up, selecting potentially necessary second-line treatment and improving the quality of life for patients diagnosed with a malignant brain tumor. The current standard-of-care (SoC) for HGG includes follow-up neuroradiological imaging to detect recurrence as early as possible and relies on several clinical, neuropathological, and radiological prognostic factors, which have limited accuracy in predicting TTR. In this study, using an in-silico analysis, we aim to improve predictive power for TTR by considering the role of (i) prognostically relevant information available through diagnostics used in the current SoC, (ii) advanced image-based information not currently part of the standard diagnostic workup, such as tumor-normal tissue interface (edge) features and quantitative data specific to biopsy positions within the tumor, and (iii) information on tumor-associated macrophages. In particular, we introduced a state-of-the-art spatio-temporal model of tumor-immune interactions, emphasizing the interplay between macrophages and glioma cells. This model serves as a synthetic reality for assessing the predictive value of various features. We generated a cohort of virtual patients based on our mathematical model. Each patient's dataset includes simulated T1Gd and Fluid-attenuated inversion recovery (FLAIR) MRI volumes. T1-weighted imaging highlights anatomical structures with high contrast, providing clear detail on brain morphology, whereas FLAIR suppresses fluid signals, improving the visualization of lesions near fluid-filled spaces, which is particularly helpful for identifying peritumoral edema. Additionally, we simulated results on macrophage density and proliferative activity, either in a specified part of the tumor, namely the tumor core or edge ("localized"), or unspecified ("non-localized"). To enhance the realism of our synthetic data, we imposed different levels of noise. Our findings reveal that macrophage density at the tumor edge contributed to a high predictive value of feature importance for the selected regression model. Moreover, there are lower MSE values for the "localized" biopsy in prediction accuracy toward recurrence post-resection compared with "non-localized" specimens in the noisy data. In conclusion, the results show that localized biopsies provided more information about tumor behavior, especially at the interface of tumor and normal tissue (Edge).
Collapse
Affiliation(s)
- Pejman Shojaee
- Center for Interdisciplinary Digital Sciences (CIDS), Department Information Services and High-Performance Computing (ZIH), Dresden University of Technology, 01062, Dresden, Germany
| | - Edwin Weinholtz
- Center for Interdisciplinary Digital Sciences (CIDS), Department Information Services and High-Performance Computing (ZIH), Dresden University of Technology, 01062, Dresden, Germany
| | - Nadine S Schaadt
- Department of Neuropathology, Institute for Pathology, Hannover Medical School, Hannover, Germany
| | - Friedrich Feuerhake
- Department of Neuropathology, Institute for Pathology, Hannover Medical School, Hannover, Germany
- Institute for Neuropathology, University Clinic Freiburg, Freiburg, Germany
| | - Haralampos Hatzikirou
- Center for Interdisciplinary Digital Sciences (CIDS), Department Information Services and High-Performance Computing (ZIH), Dresden University of Technology, 01062, Dresden, Germany.
- Mathematics Department, Khalifa University, Abu Dhabi, UAE.
| |
Collapse
|
10
|
Prado MB, Coelho BP, Iglesia RP, Alves RN, Boccacino JM, Fernandes CFL, Melo-Escobar MI, Ayyadhury S, Cruz MC, Santos TG, Beraldo FH, Fan J, Ferreira FM, Nakaya HI, Prado MAM, Prado VF, Duennwald ML, Lopes MH. Prion protein regulates invasiveness in glioblastoma stem cells. BMC Cancer 2024; 24:1539. [PMID: 39695426 DOI: 10.1186/s12885-024-13285-4] [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: 05/31/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is an aggressive brain tumor driven by glioblastoma stem cells (GSCs), which represent an appealing target for therapeutic interventions. The cellular prion protein (PrPC), a scaffold protein involved in diverse cellular processes, interacts with various membrane and extracellular matrix molecules, influencing tumor biology. Herein, we investigate the impact of PrPC expression on GBM. METHODS To address this goal, we employed CRISPR-Cas9 technology to generate PrPC knockout (KO) glioblastoma cell lines, enabling detailed loss-of-function studies. Bulk RNA sequencing followed by differentially expressed gene and pathway enrichment analyses between U87 or U251 PrPC-wild-type (WT) cells and PrPC-knockout (KO) cells were used to identify pathways regulated by PrPC. Immunofluorescence assays were used to evaluate cellular morphology and protein distribution. For assessment of protein levels, Western blot and flow cytometry assays were employed. Transwell and growth curve assays were used to determine the impact of loss-of-PrPC in GBM invasiveness and proliferation, respectively. Single-cell RNA sequencing analysis of data from patient tumors from The Cancer Genome Atlas (TCGA) and the Broad Institute of Single-Cell Data Portal were used to evaluate the correspondence between our in vitro results and patient samples. RESULTS Transcriptome analysis of PrPC-KO GBM cell lines revealed altered expression of genes associated with crucial tumor progression pathways, including migration, proliferation, and stemness. These findings were corroborated by assays that revealed impaired invasion, migration, proliferation, and self-renewal in PrPC-KO GBM cells, highlighting its critical role in sustaining tumor growth. Notably, loss-of-PrPC disrupted the expression and localization of key stemness markers, particularly CD44. Additionally, the modulation of PrPC levels through CD44 overexpression further emphasizes their regulatory role in these processes. CONCLUSIONS These findings establish PrPC as a modulator of essential molecules on the cell surface of GSCs, highlighting its potential as a therapeutic target for GBM.
Collapse
Affiliation(s)
- Mariana B Prado
- Laboratory of Neurobiology and Stem Cells, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Bárbara P Coelho
- Laboratory of Neurobiology and Stem Cells, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Rebeca P Iglesia
- Laboratory of Neurobiology and Stem Cells, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Rodrigo N Alves
- Laboratory of Neurobiology and Stem Cells, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Jacqueline M Boccacino
- Laboratory of Neurobiology and Stem Cells, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Camila F L Fernandes
- Laboratory of Neurobiology and Stem Cells, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Maria Isabel Melo-Escobar
- Laboratory of Neurobiology and Stem Cells, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Shamini Ayyadhury
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Mario C Cruz
- Core Facility to Support Research - Institute of Biomedical Sciences (CEFAP), Sao Paulo, Brazil
| | - Tiago G Santos
- Laboratory of Cell and Molecular Biology, International Research Center, A.C. Camargo Cancer Center, Sao Paulo, SP, Brazil
| | - Flávio H Beraldo
- Robarts Research Institute, Departments of Physiology and Pharmacology, Anatomy and Cell Biology, and Biochemistry, The University of Western Ontario, London, Ontario, Canada
- Schulich School of Medicine & Dentistry, Department of Pathology and Laboratory Medicine, The University of Western Ontario, London, Ontario, Canada
| | - Jue Fan
- Robarts Research Institute, Departments of Physiology and Pharmacology, Anatomy and Cell Biology, and Biochemistry, The University of Western Ontario, London, Ontario, Canada
| | - Frederico M Ferreira
- LIM50, Division of Pathology, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Helder I Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Scientific Platform Pasteur, University of São Paulo, São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Marco A M Prado
- Robarts Research Institute, Departments of Physiology and Pharmacology, Anatomy and Cell Biology, and Biochemistry, The University of Western Ontario, London, Ontario, Canada
- Schulich School of Medicine & Dentistry, Department of Pathology and Laboratory Medicine, The University of Western Ontario, London, Ontario, Canada
| | - Vania F Prado
- Robarts Research Institute, Departments of Physiology and Pharmacology, Anatomy and Cell Biology, and Biochemistry, The University of Western Ontario, London, Ontario, Canada
- Schulich School of Medicine & Dentistry, Department of Pathology and Laboratory Medicine, The University of Western Ontario, London, Ontario, Canada
| | - Martin L Duennwald
- Schulich School of Medicine & Dentistry, Department of Pathology and Laboratory Medicine, The University of Western Ontario, London, Ontario, Canada
| | - Marilene H Lopes
- Laboratory of Neurobiology and Stem Cells, Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
11
|
Knowles LM, Wolter C, Linsler S, Müller S, Urbschat S, Ketter R, Müller A, Zhou X, Qu B, Senger S, Geisel J, Schmidt T, Eichler H, Pilch J. Clotting Promotes Glioma Growth and Infiltration Through Activation of Focal Adhesion Kinase. CANCER RESEARCH COMMUNICATIONS 2024; 4:3124-3136. [PMID: 39530733 PMCID: PMC11638908 DOI: 10.1158/2767-9764.crc-24-0164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/15/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
SIGNIFICANCE High-grade gliomas are associated with intratumoral thrombosis, tumor cell necrosis, and hemorrhage. The resulting blood clot serves as an adhesive matrix for glioma cell integrins that activate FAK. Knocking down FAK with CRISPR cas9, on the other hand, is highly effective at halting GBM growth in mice.
Collapse
Affiliation(s)
- Lynn M. Knowles
- Institute of Clinical Hemostaseology and Transfusion Medicine, Saarland University and University Medical Center, Homburg, Germany
| | - Carolin Wolter
- Institute of Clinical Hemostaseology and Transfusion Medicine, Saarland University and University Medical Center, Homburg, Germany
| | - Stefan Linsler
- Department of Neurosurgery, Saarland University and University Medical Center, Homburg, Germany
| | - Simon Müller
- Department of Neurosurgery, Saarland University and University Medical Center, Homburg, Germany
| | - Steffi Urbschat
- Department of Neurosurgery, Saarland University and University Medical Center, Homburg, Germany
| | - Ralf Ketter
- Department of Neurosurgery, Saarland University and University Medical Center, Homburg, Germany
| | - Andreas Müller
- Clinic for Diagnostic and Interventional Radiology, Saarland University and University Medical Center, Homburg, Germany
| | - Xiangda Zhou
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University and University Medical Center, Homburg, Germany
| | - Bin Qu
- Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University and University Medical Center, Homburg, Germany
| | - Sebastian Senger
- Department of Neurosurgery, Saarland University and University Medical Center, Homburg, Germany
| | - Jürgen Geisel
- Clinical Chemistry and Laboratory Medicine, Saarland University and University Medical Center, Homburg, Germany
| | - Tim Schmidt
- Clinical Chemistry and Laboratory Medicine, Saarland University and University Medical Center, Homburg, Germany
| | - Hermann Eichler
- Institute of Clinical Hemostaseology and Transfusion Medicine, Saarland University and University Medical Center, Homburg, Germany
| | - Jan Pilch
- Institute of Clinical Hemostaseology and Transfusion Medicine, Saarland University and University Medical Center, Homburg, Germany
| |
Collapse
|
12
|
Chandratre S, Merenich D, Myers K, Chen B. 5-Aminolevulinic acid-mediated photodynamic therapy in combination with kinase inhibitor lapatinib enhances glioblastoma cell death. Apoptosis 2024; 29:1978-1987. [PMID: 39190205 PMCID: PMC11550286 DOI: 10.1007/s10495-024-02012-w] [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] [Accepted: 08/15/2024] [Indexed: 08/28/2024]
Abstract
5-Aminolevulinic acid (ALA) is an intraoperative imaging agent approved for protoporphyrin IX (PpIX) fluorescence-guided resection of glioblastoma (GBM). It is currently under clinical evaluation for photodynamic therapy (PDT) after the completion of GBM surgery. We previously showed that lapatinib, a clinical kinase inhibitor of epidermal growth factor receptor 1 & 2 (EGFR and HER2), enhanced PpIX fluorescence in a panel of GBM cell lines by blocking ABCG2 (ATP-binding cassette super-family G member 2)-mediated PpIX efflux, which suggests its potential for improving ALA for GBM surgery and PDT. Here we show that lapatinib enhanced PDT-induced cytotoxicity by promoting GBM cell death with the induction of apoptosis followed by necrosis. While the induction of tumor cell apoptosis was massive and rapid in the H4 cell line with no detectable Bcl-2 and a low level of Bcl-xL, it was delayed and much less in extent in A172, U-87 and U-118 cell lines with higher levels of pro-survival Bcl-2 family proteins. Lapatinib treatment alone neither reduced GBM cell viability nor had any significant effect on EGFR downstream signaling. Its enhancement of ALA-PDT was largely due to the increase of intracellular PpIX particularly in the mitochondria, resulting in the activation of mitochondria-mediated apoptosis in H4 cells. Our present study demonstrates that lapatinib inhibits ABCG2-mediated PpIX efflux and sensitizes GBM cells to ALA-PDT by inducing tumor cell death.
Collapse
Affiliation(s)
- Sharayu Chandratre
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, Saint Joseph's University, Philadelphia, PA, 19104, USA
| | - Daniel Merenich
- Department of Biology, College of Arts and Sciences, Saint Joseph's University, Philadelphia, PA, USA
| | - Kenneth Myers
- Department of Biology, College of Arts and Sciences, Saint Joseph's University, Philadelphia, PA, USA
| | - Bin Chen
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, Saint Joseph's University, Philadelphia, PA, 19104, USA.
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
13
|
Nomura N, Nagasaka S, Suzuki K, Yamamoto J. Imaging-tracked progression of primary leptomeningeal gliomatosis: A case report. Surg Neurol Int 2024; 15:411. [PMID: 39640309 PMCID: PMC11618755 DOI: 10.25259/sni_759_2024] [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: 09/07/2024] [Accepted: 10/23/2024] [Indexed: 12/07/2024] Open
Abstract
Background Primary leptomeningeal gliomatosis (PLG) is a rare neoplasm characterized by the diffuse spread of glial tumor cells throughout the leptomeninges without any evidence of a primary tumor source in the brain or spinal cord parenchyma. Here, we present a case of PLG potentially linked to prior interventional radiotherapy. Case Description The patient was a 75-year-old woman with a history of interventional radiology for a left internal carotid cavernous sinus fistula 13 years before presentation. Routine follow-up fluid-attenuated inversion recovery magnetic resonance imaging revealed a high intensity region spreading from the deep white matter of the subventricular zone (SVZ) to the insular cortex and medial temporal lobe. Subsequently, contrast-enhanced T1-weighted imaging revealed an enhanced effect consistent with extensive leptomeninges extending from the basilar cistern to the left Sylvian fissure. The patient underwent surgery, and subsequently histological examination of extracted tissue revealed a glioblastoma (GBM). Despite postoperative concurrent chemoradiotherapy and adjuvant temozolomide chemotherapy, the tumor increased in size, and the patient died 2 months postoperatively. Conclusion This case highlights the importance of careful follow-up and early therapeutic intervention in PLG, as it can be difficult to diagnose leptomeningeal lesions alone. This case also raises the possibility of radiation-induced GBM, and the criteria for diagnosis were fully met. The progression of PLG from the SVZ to the leptomeningeal site was tracked using imaging, providing valuable insights into the pattern of spread of this rare condition.
Collapse
Affiliation(s)
| | - Shohei Nagasaka
- Department of Neurosurgery, University of Occupational and Environmental Health, Kitakyusyu, Japan
| | | | | |
Collapse
|
14
|
Norollahi SE, Yousefi B, Nejatifar F, Yousefzadeh-Chabok S, Rashidy-Pour A, Samadani AA. Practical immunomodulatory landscape of glioblastoma multiforme (GBM) therapy. J Egypt Natl Canc Inst 2024; 36:33. [PMID: 39465481 DOI: 10.1186/s43046-024-00240-4] [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: 01/26/2024] [Accepted: 09/21/2024] [Indexed: 10/29/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most common harmful high-grade brain tumor with high mortality and low survival rate. Importantly, besides routine diagnostic and therapeutic methods, modern and useful practical techniques are urgently needed for this serious malignancy. Correspondingly, the translational medicine focusing on genetic and epigenetic profiles of glioblastoma, as well as the immune framework and brain microenvironment, based on these challenging findings, indicates that key clinical interventions include immunotherapy, such as immunoassay, oncolytic viral therapy, and chimeric antigen receptor T (CAR T) cell therapy, which are of great importance in both diagnosis and therapy. Relatively, vaccine therapy reflects the untapped confidence to enhance GBM outcomes. Ongoing advances in immunotherapy, which utilizes different methods to regenerate or modify the resistant body for cancer therapy, have revealed serious results with many different problems and difficulties for patients. Safe checkpoint inhibitors, adoptive cellular treatment, cellular and peptide antibodies, and other innovations give researchers an endless cluster of instruments to plan profoundly in personalized medicine and the potential for combination techniques. In this way, antibodies that block immune checkpoints, particularly those that target the program death 1 (PD-1)/PD-1 (PD-L1) ligand pathway, have improved prognosis in a wide range of diseases. However, its use in combination with chemotherapy, radiation therapy, or monotherapy is ineffective in treating GBM. The purpose of this review is to provide an up-to-date overview of the translational elements concentrating on the immunotherapeutic field of GBM alongside describing the molecular mechanism involved in GBM and related signaling pathways, presenting both historical perspectives and future directions underlying basic and clinical practice.
Collapse
Affiliation(s)
- Seyedeh Elham Norollahi
- Cancer Research Center and, Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | - Bahman Yousefi
- Cancer Research Center and, Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | - Fatemeh Nejatifar
- Department of Hematology and Oncology, School of Medicine, Razi Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Shahrokh Yousefzadeh-Chabok
- Guilan Road Trauma Research Center, Trauma Institute, Guilan University of Medical Sciences, Rasht, Iran
- , Rasht, Iran
| | - Ali Rashidy-Pour
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran.
| | - Ali Akbar Samadani
- Guilan Road Trauma Research Center, Trauma Institute, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
15
|
Bartos LM, Quach S, Zenatti V, Kirchleitner SV, Blobner J, Wind-Mark K, Kolabas ZI, Ulukaya S, Holzgreve A, Ruf VC, Kunze LH, Kunte ST, Hoermann L, Härtel M, Park HE, Groß M, Franzmeier N, Zatcepin A, Zounek A, Kaiser L, Riemenschneider MJ, Perneczky R, Rauchmann BS, Stöcklein S, Ziegler S, Herms J, Ertürk A, Tonn JC, Thon N, von Baumgarten L, Prestel M, Tahirovic S, Albert NL, Brendel M. Remote Neuroinflammation in Newly Diagnosed Glioblastoma Correlates with Unfavorable Clinical Outcome. Clin Cancer Res 2024; 30:4618-4634. [PMID: 39150564 PMCID: PMC11474166 DOI: 10.1158/1078-0432.ccr-24-1563] [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: 05/22/2024] [Revised: 07/15/2024] [Accepted: 08/14/2024] [Indexed: 08/17/2024]
Abstract
PURPOSE Current therapy strategies still provide only limited success in the treatment of glioblastoma, the most frequent primary brain tumor in adults. In addition to the characterization of the tumor microenvironment, global changes in the brain of patients with glioblastoma have been described. However, the impact and molecular signature of neuroinflammation distant of the primary tumor site have not yet been thoroughly elucidated. EXPERIMENTAL DESIGN We performed translocator protein (TSPO)-PET in patients with newly diagnosed glioblastoma (n = 41), astrocytoma WHO grade 2 (n = 7), and healthy controls (n = 20) and compared TSPO-PET signals of the non-lesion (i.e., contralateral) hemisphere. Back-translation into syngeneic SB28 glioblastoma mice was used to characterize Pet alterations on a cellular level. Ultimately, multiplex gene expression analyses served to profile immune cells in remote brain. RESULTS Our study revealed elevated TSPO-PET signals in contralateral hemispheres of patients with newly diagnosed glioblastoma compared to healthy controls. Contralateral TSPO was associated with persisting epileptic seizures and shorter overall survival independent of the tumor phenotype. Back-translation into syngeneic glioblastoma mice pinpointed myeloid cells as the predominant source of contralateral TSPO-PET signal increases and identified a complex immune signature characterized by myeloid cell activation and immunosuppression in distant brain regions. CONCLUSIONS Neuroinflammation within the contralateral hemisphere can be detected with TSPO-PET imaging and associates with poor outcome in patients with newly diagnosed glioblastoma. The molecular signature of remote neuroinflammation promotes the evaluation of immunomodulatory strategies in patients with detrimental whole brain inflammation as reflected by high TSPO expression.
Collapse
Affiliation(s)
- Laura M. Bartos
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Stefanie Quach
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany.
| | - Valerio Zenatti
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany.
| | | | - Jens Blobner
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany.
| | - Karin Wind-Mark
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Zeynep Ilgin Kolabas
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Munich, Germany.
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
- Graduate School of Systemic Neurosciences (GSN), Munich, Germany.
| | - Selin Ulukaya
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Munich, Germany.
- Faculty of Biology, Master of Science Program in Molecular and Cellular Biology, Ludwig-Maximilians-Universität München, Planegg, Germany.
| | - Adrien Holzgreve
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Viktoria C. Ruf
- Center for Neuropathology and Prion Research, University Hospital, LMU Munich, Munich, Germany.
| | - Lea H. Kunze
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Sebastian T. Kunte
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Leonie Hoermann
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Marlies Härtel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Ha Eun Park
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Mattes Groß
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
| | - Artem Zatcepin
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany.
| | - Adrian Zounek
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Lena Kaiser
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | | | - Robert Perneczky
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), University of Munich, Munich, Germany.
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany.
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kingdom.
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, United Kingdom.
| | | | - Sophia Stöcklein
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany.
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
| | - Jochen Herms
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany.
- Center for Neuropathology and Prion Research, University Hospital, LMU Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), University of Munich, Munich, Germany.
| | - Ali Ertürk
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Munich, Germany.
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
- Graduate School of Systemic Neurosciences (GSN), Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), University of Munich, Munich, Germany.
| | - Joerg C. Tonn
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany.
| | - Niklas Thon
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Louisa von Baumgarten
- Department of Neurosurgery, University Hospital, LMU Munich, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany.
| | - Matthias Prestel
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany.
| | - Sabina Tahirovic
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany.
| | - Nathalie L. Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany.
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.
- DZNE – German Center for Neurodegenerative Diseases, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Munich Cluster for Systems Neurology (SyNergy), University of Munich, Munich, Germany.
| |
Collapse
|
16
|
Rodriguez SMB, Tataranu LG, Kamel A, Turliuc S, Rizea RE, Dricu A. Glioblastoma and Immune Checkpoint Inhibitors: A Glance at Available Treatment Options and Future Directions. Int J Mol Sci 2024; 25:10765. [PMID: 39409094 PMCID: PMC11477435 DOI: 10.3390/ijms251910765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
Glioblastoma is known to be one of the most aggressive and fatal human cancers, with a poor prognosis and resistance to standard treatments. In the last few years, many solid tumor treatments have been revolutionized with the help of immunotherapy. However, this type of treatment has failed to improve the results in glioblastoma patients. Effective immunotherapeutic strategies may be developed after understanding how glioblastoma achieves tumor-mediated immune suppression in both local and systemic landscapes. Biomarkers may help identify patients most likely to benefit from this type of treatment. In this review, we discuss the use of immunotherapy in glioblastoma, with an emphasis on immune checkpoint inhibitors and the factors that influence clinical response. A Pubmed data search was performed for all existing information regarding immune checkpoint inhibitors used for the treatment of glioblastoma. All data evaluating the ongoing clinical trials involving the use of ICIs either as monotherapy or in combination with other drugs was compiled and analyzed.
Collapse
Affiliation(s)
- Silvia Mara Baez Rodriguez
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
| | - Ligia Gabriela Tataranu
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
- Neurosurgical Department, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania
| | - Amira Kamel
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
| | - Serban Turliuc
- Medical Department, University of Medicine and Pharmacy “G. T. Popa”, 700115 Iasi, Romania;
| | - Radu Eugen Rizea
- Neurosurgical Department, Bagdasar-Arseni Clinical Emergency Hospital, 041915 Bucharest, Romania; (S.M.B.R.); (A.K.); (R.E.R.)
- Neurosurgical Department, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania
| | - Anica Dricu
- Biochemistry Department, Carol Davila University of Medicine and Pharmacy, 020022 Bucharest, Romania;
| |
Collapse
|
17
|
Zhang B, Wang W, Song Y, Chen H, Lin X, Chen J, Chen Y, Huang J, Li D, Wu S. Exploring the Mechanism of Sempervirine Inhibiting Glioblastoma Invasion Based on Network Pharmacology and Bioinformatics. Pharmaceuticals (Basel) 2024; 17:1318. [PMID: 39458959 PMCID: PMC11510114 DOI: 10.3390/ph17101318] [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: 08/26/2024] [Revised: 09/19/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Invasion is an important characteristic of the malignancy of glioblastoma (GBM) and a significant prognostic factor. Sempervirine (SPV), a yohimbine-type alkaloid, has been proven to inhibit GBM cells proliferation in previous research and found to have a potential effect in anti-invasion, but its mechanism of anti-invasion is still unknown. Methods: To explore its pharmacodynamics in inhibiting GBM cell invasion in this study, we combined network pharmacology and bioinformatics to comprehensive exploratory analysis of SPV and verified the mechanism in vitro. Results: Firstly, targets of SPV and invasion-related genes were collected from public databases. Moreover, GBM samples were obtained to analyze differentially expressed genes (DEGs) from The Cancer Genome Atlas (TCGA). Then, the relevant targets of SPV inhibiting GBM invasion (SIGI) were obtained through the intersection of the three gene sets. Further, GO and KEGG analysis showed that the targets of SIGI were heavily enriched in the AKT signaling pathway. Subsequently, based on the method of machine learning, a clinical prognostic model of the relevant targets of SIGI was constructed using GBM samples from TCGA and the Gene Expression Omnibus (GEO). A four-genes model (DUSP6, BMP2, MMP2, and MMP13) was successfully constructed, and Vina Scores of MMP2 and MMP13 in molecular docking were higher, which may be the main targets of SIGI. Then, the effect of SIGI was confirmed via functional experiments on invasion, migration, and adhesion assay, and the effect involved changes in the expressions of p-AKT, MMP2 and MMP13. Finally, combined with AKT activator (SC79) and inhibitor (MK2206), we further confirmed that SPV inhibits GBM invasion through AKT phosphorylation. Conclusions: This study provides valuable and an expected point of view into the regulation of AKT phosphorylation and inhibition of GBM invasion by SPV.
Collapse
Affiliation(s)
- Bingqiang Zhang
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (B.Z.); (Y.S.); (H.C.); (X.L.); (J.C.); (Y.C.)
| | - Wenyi Wang
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China;
| | - Yu Song
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (B.Z.); (Y.S.); (H.C.); (X.L.); (J.C.); (Y.C.)
| | - Huixian Chen
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (B.Z.); (Y.S.); (H.C.); (X.L.); (J.C.); (Y.C.)
| | - Xinxin Lin
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (B.Z.); (Y.S.); (H.C.); (X.L.); (J.C.); (Y.C.)
| | - Jingjing Chen
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (B.Z.); (Y.S.); (H.C.); (X.L.); (J.C.); (Y.C.)
| | - Ying Chen
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (B.Z.); (Y.S.); (H.C.); (X.L.); (J.C.); (Y.C.)
| | - Jinfang Huang
- Fuzhou First General Hospital, Fuzhou 350009, China;
| | - Desen Li
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (B.Z.); (Y.S.); (H.C.); (X.L.); (J.C.); (Y.C.)
| | - Shuisheng Wu
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (B.Z.); (Y.S.); (H.C.); (X.L.); (J.C.); (Y.C.)
| |
Collapse
|
18
|
Ji X, Cheng J, Su J, Wen R, Zhang Q, Liu G, Peng Y, Mao J. PTPN7 mediates macrophage-polarization and determines immunotherapy in gliomas: A single-cell sequencing analysis. ENVIRONMENTAL TOXICOLOGY 2024; 39:4562-4580. [PMID: 38581214 DOI: 10.1002/tox.24259] [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: 01/13/2024] [Revised: 03/09/2024] [Accepted: 03/23/2024] [Indexed: 04/08/2024]
Abstract
BACKGROUND Protein tyrosine phosphatase non-receptor type 7 (PTPN7) is a signaling molecule that regulates a multitude of cellular processes, spanning cell proliferation, cellular differentiation, the mitotic cycle, and oncogenic metamorphosis. However, the characteristic of PTPN7 in the glioma microenvironment has yet to be elucidated. METHODS The prognostic value, genomic features, immune characteristics, chemotherapy prediction, and immunotherapy prediction of PTPN7 were systematically explored at the bulk sequencing level. The cell evolution trajectory, cell communication pattern, and cell metabolic activity related to PTPN7 were systematically explored at the single-cell sequencing level. HMC3 and M0 cells were cocultured with U251 and T98G cells, and flow cytometry was carried out to investigate the polarization of HMC3 and M0. Transwell assay and CCK-8 assay were performed to explore the migration and proliferation activity of U251 and T98G. RESULTS The expression level of PTPN7 is significantly elevated in glioma and indicates malignant features. PTPN7 expression predicts worse prognosis of glioma patients. PTPN7 is associated with genome alteration and immune infiltration. Besides, PTPN7 plays a crucial role in modulating metabolic and immunogenic processes, particularly by influencing the activity of microglia and macrophages through multiple signaling pathways involved in cellular communication. Specifically, PTPN7 actively mediates inflammation-resolving-polarization of macrophages and microglia and protects glioma from immune attack. PTPN7 could also predict the response of immunotherapy. CONCLUSIONS PTPN7 is critically involved in inflammation-resolving-polarization mediated by macrophage and microglia and promotes the immune escape of glioma cells.
Collapse
Affiliation(s)
- Xiang Ji
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jingsong Cheng
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jing Su
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Rong Wen
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Qi Zhang
- Department of Neurosurgery, Tongnan Hospital of TCM, Chongqing, China
| | - Guodong Liu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yun Peng
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jinning Mao
- Health Management Center, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
19
|
Zeng X, Ropper AE, Aljuboori Z, Yu D, Teng TW, Kabatas S, Usuga E, Anderson JE, Teng YD. Concurrent Oncolysis and Neurolesion Repair by Dual Gene-Engineered hNSCs in an Experimental Model of Intraspinal Cord Glioblastoma. Cells 2024; 13:1522. [PMID: 39329707 PMCID: PMC11429792 DOI: 10.3390/cells13181522] [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: 06/13/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 09/28/2024] Open
Abstract
Intramedullary spinal cord glioblastoma (ISCG) is lethal due to lack of effective treatment. We previously established a rat C6-ISCG model and the antitumor effect of F3.CD-TK, an hNSC line expressing CD and TK, via producing cytocidal 5FU and GCV-TP. However, the neurotherapeutic potential of this hNSC approach has remained uninvestigated. Here for the first time, cultured F3.CD-TK cells were found to have a markedly higher oncolytic effect, which was GJIC-dependent, and BDNF expression but less VEGF secretion than F3.CD. In Rowett athymic rats, F3.CD-TK (1.5 × 106 cells/10 µL × 2), injected near C6-ISCG (G55 seeding 7 days earlier: 10 K/each) and followed by q.d. (×5/each repeat; i.p.) of 5FC (500 mg/kg/5 mL/day) and GCV (25 mg/kg/1 mL/day), robustly mitigated cardiorespiratory, locomotor, and sensory deficits to improve neurofunction and overall survival compared to animals receiving either F3.CD or F3.CD-TK+F3.CD debris formula. The F3.CD-TK regimen exerted greater tumor penetration and neural inflammation/immune modulation, reshaped C6-ISCG topology to increase the tumor's surface area/volume ratio to spare/repair host axons (e.g., vGlut1+ neurites), and had higher post-prodrug donor self-clearance. The multimodal data and mechanistic leads from this proof-of-principle study suggest that the overall stronger anti-ISCG benefit of our hNSC-based GDEPT is derived from its concurrent oncolytic and neurotherapeutic effects.
Collapse
Affiliation(s)
- Xiang Zeng
- Department of Physical Medicine and Rehabilitation, Harvard Medical School and Spaulding Rehabilitation Hospital, Boston, MA 02129, USA
- Department of Neurosurgery, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| | - Alexander E. Ropper
- Department of Neurosurgery, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| | - Zaid Aljuboori
- Department of Neurosurgery, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| | - Dou Yu
- Department of Physical Medicine and Rehabilitation, Harvard Medical School and Spaulding Rehabilitation Hospital, Boston, MA 02129, USA
- Department of Neurosurgery, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| | | | - Serdar Kabatas
- Department of Neurosurgery, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| | - Esteban Usuga
- Department of Physical Medicine and Rehabilitation, Harvard Medical School and Spaulding Rehabilitation Hospital, Boston, MA 02129, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| | - Jamie E. Anderson
- Department of Physical Medicine and Rehabilitation, Harvard Medical School and Spaulding Rehabilitation Hospital, Boston, MA 02129, USA
- Department of Neurosurgery, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| | - Yang D. Teng
- Department of Physical Medicine and Rehabilitation, Harvard Medical School and Spaulding Rehabilitation Hospital, Boston, MA 02129, USA
- Department of Neurosurgery, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
- Laboratory of SCI, Stem Cell, and Recovery Neurobiology Research, Department of Physical Medicine and Rehabilitation, Spaulding Rehabilitation Hospital Network, Mass General Brigham, and Harvard Medical School, Boston, MA 02129, USA
| |
Collapse
|
20
|
Papacocea SI, Vrinceanu D, Dumitru M, Manole F, Serboiu C, Papacocea MT. Molecular Profile as an Outcome Predictor in Glioblastoma along with MRI Features and Surgical Resection: A Scoping Review. Int J Mol Sci 2024; 25:9714. [PMID: 39273661 PMCID: PMC11395592 DOI: 10.3390/ijms25179714] [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: 07/25/2024] [Revised: 09/06/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive malignant tumors of the brain. We queried PubMed for articles about molecular predictor markers in GBM. This scoping review aims to analyze the most important outcome predictors in patients with GBM and to compare these factors in terms of absolute months of survival benefit and percentages. Performing a gross total resection for patients with GBM undergoing optimal chemo- and radiotherapy provides a significant benefit in overall survival compared to those patients who received a subtotal or partial resection. However, compared to IDH-Wildtype GBMs, patients with IDH-Mutant 1/2 GBMs have an increased survival. MGMT promoter methylation status is another strong outcome predictor for patients with GBM. In the reviewed literature, patients with methylated MGMT promoter lived approximately 50% to 90% longer than those with an unmethylated MGMT gene promoter. Moreover, KPS is an important predictor of survival and quality of life, demonstrating that we should refrain from aggressive surgery in important brain areas. As new therapies (such as TTFs) emerge, we are optimistic that the overall median survival will increase, even for IDH-Wildtype GBMs. In conclusion, molecular profiles are stronger outcome predictors than the extent of neurosurgical resection for GBM.
Collapse
Affiliation(s)
- Serban Iancu Papacocea
- Neurosurgery Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (S.I.P.); (M.T.P.)
| | - Daniela Vrinceanu
- ENT Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Mihai Dumitru
- ENT Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Felicia Manole
- ENT Department, Faculty of Medicine, University of Oradea, 410073 Oradea, Romania;
| | - Crenguta Serboiu
- Cellular Biology and Histology Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Marius Toma Papacocea
- Neurosurgery Department, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (S.I.P.); (M.T.P.)
| |
Collapse
|
21
|
Wang J, Shen S, You J, Wang Z, Li Y, Chen Y, Tuo Y, Chen D, Yu H, Zhang J, Wang F, Pang X, Xiao Z, Lan Q, Wang Y. PRMT6 facilitates EZH2 protein stability by inhibiting TRAF6-mediated ubiquitination degradation to promote glioblastoma cell invasion and migration. Cell Death Dis 2024; 15:524. [PMID: 39043634 PMCID: PMC11266590 DOI: 10.1038/s41419-024-06920-2] [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: 04/16/2024] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024]
Abstract
Invasion and migration are the key hallmarks of cancer, and aggressive growth is a major factor contributing to treatment failure and poor prognosis in glioblastoma. Protein arginine methyltransferase 6 (PRMT6), as an epigenetic regulator, has been confirmed to promote the malignant proliferation of glioblastoma cells in previous studies. However, the effects of PRMT6 on glioblastoma cell invasion and migration and its underlying mechanisms remain elusive. Here, we report that PRMT6 functions as a driver element for tumor cell invasion and migration in glioblastoma. Bioinformatics analysis and glioma sample detection results demonstrated that PRMT6 is highly expressed in mesenchymal subtype or invasive gliomas, and is significantly negatively correlated with their prognosis. Inhibition of PRMT6 (using PRMT6 shRNA or inhibitor EPZ020411) reduces glioblastoma cell invasion and migration in vitro, whereas overexpression of PRMT6 produces opposite effects. Then, we identified that PRMT6 maintains the protein stability of EZH2 by inhibiting the degradation of EZH2 protein, thereby mediating the invasion and migration of glioblastoma cells. Further mechanistic investigations found that PRMT6 inhibits the transcription of TRAF6 by activating the histone methylation mark (H3R2me2a), and reducing the interaction between TRAF6 and EZH2 to enhance the protein stability of EZH2 in glioblastoma cells. Xenograft tumor assay and HE staining results showed that the expression of PRMT6 could promote the invasion of glioblastoma cells in vivo, the immunohistochemical staining results of mouse brain tissue tumor sections also confirmed the regulatory relationship between PRMT6, TRAF6, and EZH2. Our findings illustrate that PRMT6 suppresses TRAF6 transcription via H3R2me2a to enhance the protein stability of EZH2 to facilitate glioblastoma cell invasion and migration. Blocking the PRMT6-TRAF6-EZH2 axis is a promising strategy for inhibiting glioblastoma cell invasion and migration.
Collapse
Affiliation(s)
- Ji Wang
- Department of Neurosurgery, Institute of Neuroscience, The Second Affiliated Hospital, Guangzhou Medical University, 510260, Guangzhou, China.
| | - Shiquan Shen
- Department of Neurosurgery, Institute of Neuroscience, The Second Affiliated Hospital, Guangzhou Medical University, 510260, Guangzhou, China
| | - Jian You
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, 646000, Luzhou, China
| | - Zhaotao Wang
- Department of Neurosurgery, Institute of Neuroscience, The Second Affiliated Hospital, Guangzhou Medical University, 510260, Guangzhou, China
| | - Yan Li
- Department of Cardiology, The First Affiliated Hospital of University of Science and Technology of China, 230001, Hefei, China
| | - Yanming Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, 215004, Suzhou, China
| | - Yonghua Tuo
- Department of Neurosurgery, Institute of Neuroscience, The Second Affiliated Hospital, Guangzhou Medical University, 510260, Guangzhou, China
| | - Danmin Chen
- Department of Neurosurgery, Institute of Neuroscience, The Second Affiliated Hospital, Guangzhou Medical University, 510260, Guangzhou, China
| | - Haoming Yu
- Department of Neurosurgery, Institute of Neuroscience, The Second Affiliated Hospital, Guangzhou Medical University, 510260, Guangzhou, China
| | - Jingbo Zhang
- Department of Neurosurgery, Institute of Neuroscience, The Second Affiliated Hospital, Guangzhou Medical University, 510260, Guangzhou, China
| | - Fangran Wang
- Department of Neurosurgery, Institute of Neuroscience, The Second Affiliated Hospital, Guangzhou Medical University, 510260, Guangzhou, China
| | - Xiao Pang
- Department of Neurosurgery, Institute of Neuroscience, The Second Affiliated Hospital, Guangzhou Medical University, 510260, Guangzhou, China
| | - Zongyu Xiao
- Department of Neurosurgery, The Fourth Affiliated Hospital of Soochow University, 215124, Suzhou, China.
| | - Qing Lan
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, 215004, Suzhou, China.
| | - Yezhong Wang
- Department of Neurosurgery, Institute of Neuroscience, The Second Affiliated Hospital, Guangzhou Medical University, 510260, Guangzhou, China.
| |
Collapse
|
22
|
Boelders SM, De Baene W, Postma E, Gehring K, Ong LL. Predicting Cognitive Functioning for Patients with a High-Grade Glioma: Evaluating Different Representations of Tumor Location in a Common Space. Neuroinformatics 2024; 22:329-352. [PMID: 38900230 PMCID: PMC11329426 DOI: 10.1007/s12021-024-09671-9] [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: 05/31/2024] [Indexed: 06/21/2024]
Abstract
Cognitive functioning is increasingly considered when making treatment decisions for patients with a brain tumor in view of a personalized onco-functional balance. Ideally, one can predict cognitive functioning of individual patients to make treatment decisions considering this balance. To make accurate predictions, an informative representation of tumor location is pivotal, yet comparisons of representations are lacking. Therefore, this study compares brain atlases and principal component analysis (PCA) to represent voxel-wise tumor location. Pre-operative cognitive functioning was predicted for 246 patients with a high-grade glioma across eight cognitive tests while using different representations of voxel-wise tumor location as predictors. Voxel-wise tumor location was represented using 13 different frequently-used population average atlases, 13 randomly generated atlases, and 13 representations based on PCA. ElasticNet predictions were compared between representations and against a model solely using tumor volume. Preoperative cognitive functioning could only partly be predicted from tumor location. Performances of different representations were largely similar. Population average atlases did not result in better predictions compared to random atlases. PCA-based representation did not clearly outperform other representations, although summary metrics indicated that PCA-based representations performed somewhat better in our sample. Representations with more regions or components resulted in less accurate predictions. Population average atlases possibly cannot distinguish between functionally distinct areas when applied to patients with a glioma. This stresses the need to develop and validate methods for individual parcellations in the presence of lesions. Future studies may test if the observed small advantage of PCA-based representations generalizes to other data.
Collapse
Affiliation(s)
- S M Boelders
- Department of Neurosurgery, Elisabeth-TweeSteden Hospital, Tilburg, The Netherlands
- Department of Cognitive Sciences and AI, Tilburg University, Tilburg, The Netherlands
| | - W De Baene
- Department of Cognitive Neuropsychology, Tilburg University Tilburg, Warandelaan 2, P. O. Box 90153, Tilburg, 5000 LE, The Netherlands
| | - E Postma
- Department of Cognitive Sciences and AI, Tilburg University, Tilburg, The Netherlands
| | - K Gehring
- Department of Neurosurgery, Elisabeth-TweeSteden Hospital, Tilburg, The Netherlands.
- Department of Cognitive Neuropsychology, Tilburg University Tilburg, Warandelaan 2, P. O. Box 90153, Tilburg, 5000 LE, The Netherlands.
| | - L L Ong
- Department of Cognitive Sciences and AI, Tilburg University, Tilburg, The Netherlands
| |
Collapse
|
23
|
Fields BKK, Calabrese E, Mongan J, Cha S, Hess CP, Sugrue LP, Chang SM, Luks TL, Villanueva-Meyer JE, Rauschecker AM, Rudie JD. The University of California San Francisco Adult Longitudinal Post-Treatment Diffuse Glioma MRI Dataset. Radiol Artif Intell 2024; 6:e230182. [PMID: 38864741 PMCID: PMC11294954 DOI: 10.1148/ryai.230182] [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: 05/23/2023] [Revised: 05/13/2024] [Accepted: 05/31/2024] [Indexed: 06/13/2024]
Abstract
Supplemental material is available for this article.
Collapse
Affiliation(s)
- Brandon K. K. Fields
- From the Center for Intelligent Imaging, Department of Radiology
& Biomedical Imaging (B.K.K.F., E.C., J.M., S.C., C.P.H., L.P.S., T.L.L.,
J.E.V.M., A.M.R., J.D.R.), and Division of Neuro-Oncology, Department of
Neurologic Surgery (S.M.C.), University of California San Francisco, 513
Parnassus Ave, Suite S-261D, Box 0628, San Francisco, CA 94143
| | | | - John Mongan
- From the Center for Intelligent Imaging, Department of Radiology
& Biomedical Imaging (B.K.K.F., E.C., J.M., S.C., C.P.H., L.P.S., T.L.L.,
J.E.V.M., A.M.R., J.D.R.), and Division of Neuro-Oncology, Department of
Neurologic Surgery (S.M.C.), University of California San Francisco, 513
Parnassus Ave, Suite S-261D, Box 0628, San Francisco, CA 94143
| | - Soonmee Cha
- From the Center for Intelligent Imaging, Department of Radiology
& Biomedical Imaging (B.K.K.F., E.C., J.M., S.C., C.P.H., L.P.S., T.L.L.,
J.E.V.M., A.M.R., J.D.R.), and Division of Neuro-Oncology, Department of
Neurologic Surgery (S.M.C.), University of California San Francisco, 513
Parnassus Ave, Suite S-261D, Box 0628, San Francisco, CA 94143
| | - Christopher P. Hess
- From the Center for Intelligent Imaging, Department of Radiology
& Biomedical Imaging (B.K.K.F., E.C., J.M., S.C., C.P.H., L.P.S., T.L.L.,
J.E.V.M., A.M.R., J.D.R.), and Division of Neuro-Oncology, Department of
Neurologic Surgery (S.M.C.), University of California San Francisco, 513
Parnassus Ave, Suite S-261D, Box 0628, San Francisco, CA 94143
| | - Leo P. Sugrue
- From the Center for Intelligent Imaging, Department of Radiology
& Biomedical Imaging (B.K.K.F., E.C., J.M., S.C., C.P.H., L.P.S., T.L.L.,
J.E.V.M., A.M.R., J.D.R.), and Division of Neuro-Oncology, Department of
Neurologic Surgery (S.M.C.), University of California San Francisco, 513
Parnassus Ave, Suite S-261D, Box 0628, San Francisco, CA 94143
| | - Susan M. Chang
- From the Center for Intelligent Imaging, Department of Radiology
& Biomedical Imaging (B.K.K.F., E.C., J.M., S.C., C.P.H., L.P.S., T.L.L.,
J.E.V.M., A.M.R., J.D.R.), and Division of Neuro-Oncology, Department of
Neurologic Surgery (S.M.C.), University of California San Francisco, 513
Parnassus Ave, Suite S-261D, Box 0628, San Francisco, CA 94143
| | - Tracy L. Luks
- From the Center for Intelligent Imaging, Department of Radiology
& Biomedical Imaging (B.K.K.F., E.C., J.M., S.C., C.P.H., L.P.S., T.L.L.,
J.E.V.M., A.M.R., J.D.R.), and Division of Neuro-Oncology, Department of
Neurologic Surgery (S.M.C.), University of California San Francisco, 513
Parnassus Ave, Suite S-261D, Box 0628, San Francisco, CA 94143
| | - Javier E. Villanueva-Meyer
- From the Center for Intelligent Imaging, Department of Radiology
& Biomedical Imaging (B.K.K.F., E.C., J.M., S.C., C.P.H., L.P.S., T.L.L.,
J.E.V.M., A.M.R., J.D.R.), and Division of Neuro-Oncology, Department of
Neurologic Surgery (S.M.C.), University of California San Francisco, 513
Parnassus Ave, Suite S-261D, Box 0628, San Francisco, CA 94143
| | | | | |
Collapse
|
24
|
Michelucci A, Catacuzzeno L. Piezo1, the new actor in cell volume regulation. Pflugers Arch 2024; 476:1023-1039. [PMID: 38581527 PMCID: PMC11166825 DOI: 10.1007/s00424-024-02951-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: 01/11/2024] [Revised: 02/29/2024] [Accepted: 03/20/2024] [Indexed: 04/08/2024]
Abstract
All animal cells control their volume through a complex set of mechanisms, both to counteract osmotic perturbations of the environment and to enable numerous vital biological processes, such as proliferation, apoptosis, and migration. The ability of cells to adjust their volume depends on the activity of ion channels and transporters which, by moving K+, Na+, and Cl- ions across the plasma membrane, generate the osmotic gradient that drives water in and out of the cell. In 2010, Patapoutian's group identified a small family of evolutionarily conserved, Ca2+-permeable mechanosensitive channels, Piezo1 and Piezo2, as essential components of the mechanically activated current that mediates mechanotransduction in vertebrates. Piezo1 is expressed in several tissues and its opening is promoted by a wide range of mechanical stimuli, including membrane stretch/deformation and osmotic stress. Piezo1-mediated Ca2+ influx is used by the cell to convert mechanical forces into cytosolic Ca2+ signals that control diverse cellular functions such as migration and cell death, both dependent on changes in cell volume and shape. The crucial role of Piezo1 in the regulation of cell volume was first demonstrated in erythrocytes, which need to reduce their volume to pass through narrow capillaries. In HEK293 cells, increased expression of Piezo1 was found to enhance the regulatory volume decrease (RVD), the process whereby the cell re-establishes its original volume after osmotic shock-induced swelling, and it does so through Ca2+-dependent modulation of the volume-regulated anion channels. More recently we reported that Piezo1 controls the RVD in glioblastoma cells via the modulation of Ca2+-activated K+ channels. To date, however, the mechanisms through which this mechanosensitive channel controls cell volume and maintains its homeostasis have been poorly investigated and are still far from being understood. The present review aims to provide a broad overview of the literature discussing the recent advances on this topic.
Collapse
Affiliation(s)
- A Michelucci
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123, Perugia, Italy.
| | - L Catacuzzeno
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123, Perugia, Italy.
| |
Collapse
|
25
|
Wang L, Li X, Xu C, Wang D, Ma C, Wang Z, Li Y, Li Z. Unveiling novel cell clusters and biomarkers in glioblastoma and its peritumoral microenvironment at the single-cell perspective. J Transl Med 2024; 22:551. [PMID: 38851695 PMCID: PMC11162569 DOI: 10.1186/s12967-024-05313-5] [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: 02/20/2024] [Accepted: 05/20/2024] [Indexed: 06/10/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a highly heterogeneous, recurrent and aggressively invasive primary malignant brain tumor. The heterogeneity of GBM results in poor targeted therapy. Therefore, the aim of this study is to depict the cellular landscape of GBM and its peritumor from a single-cell perspective. Discovering new cell subtypes and biomarkers, and providing a theoretical basis for precision therapy. METHODS We collected 8 tissue samples from 4 GBM patients to perform 10 × single-cell transcriptome sequencing. Quality control and filtering of data by Seurat package for clustering. Inferring copy number variations to identify malignant cells via the infercnv package. Functional enrichment analysis was performed by GSVA and clusterProfiler packages. STRING database and Cytoscape software were used to construct protein interaction networks. Inferring transcription factors by pySCENIC. Building cell differentiation trajectories via the monocle package. To infer intercellular communication networks by CellPhoneDB software. RESULTS We observed that the tumor microenvironment (TME) varies among different locations and different GBM patients. We identified a proliferative cluster of oligodendrocytes with high expression of mitochondrial genes. We also identified two clusters of myeloid cells, one primarily located in the peritumor exhibiting an M1 phenotype with elevated TNFAIP8L3 expression, and another in the tumor and peritumor showing a proliferative tendency towards an M2 phenotype with increased DTL expression. We identified XIST, KCNH7, SYT1 and DIAPH3 as potential factors associated with the proliferation of malignant cells in GBM. CONCLUSIONS These biomarkers and cell clusters we discovered may serve as targets for treatment. Targeted drugs developed against these biomarkers and cell clusters may enhance treatment efficacy, optimize immune therapy strategies, and improve the response rates of GBM patients to immunotherapy. Our findings provide a theoretical basis for the development of individualized treatment and precision medicine for GBM, which may be used to improve the survival of GBM patients.
Collapse
Affiliation(s)
- Liping Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Xinyi Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Chengshi Xu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Danwen Wang
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Chao Ma
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China
| | - Zefen Wang
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, Hubei, China
| | - Yirong Li
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China.
| | - Zhiqiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China.
- Brain Glioma Center, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, 430071, Hubei, China.
| |
Collapse
|
26
|
Aebisher D, Rogóż K, Myśliwiec A, Dynarowicz K, Wiench R, Cieślar G, Kawczyk-Krupka A, Bartusik-Aebisher D. The use of photodynamic therapy in medical practice. Front Oncol 2024; 14:1373263. [PMID: 38803535 PMCID: PMC11129581 DOI: 10.3389/fonc.2024.1373263] [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: 01/19/2024] [Accepted: 04/16/2024] [Indexed: 05/29/2024] Open
Abstract
Cancer therapy, especially for tumors near sensitive areas, demands precise treatment. This review explores photodynamic therapy (PDT), a method leveraging photosensitizers (PS), specific wavelength light, and oxygen to target cancer effectively. Recent advancements affirm PDT's efficacy, utilizing ROS generation to induce cancer cell death. With a history spanning over decades, PDT's dynamic evolution has expanded its application across dermatology, oncology, and dentistry. This review aims to dissect PDT's principles, from its inception to contemporary medical applications, highlighting its role in modern cancer treatment strategies.
Collapse
Affiliation(s)
- David Aebisher
- Department of Photomedicine and Physical Chemistry, Medical College of The Rzeszów University, Rzeszów, Poland
| | - Kacper Rogóż
- English Division Science Club, Medical College of The Rzeszów University, Rzeszów, Poland
| | - Angelika Myśliwiec
- Center for Innovative Research in Medical and Natural Sciences, Medical College of The University of Rzeszów, Rzeszów, Poland
| | - Klaudia Dynarowicz
- Center for Innovative Research in Medical and Natural Sciences, Medical College of The University of Rzeszów, Rzeszów, Poland
| | - Rafał Wiench
- Department of Periodontal Diseases and Oral Mucosa Diseases, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Zabrze, Poland
| | - Grzegorz Cieślar
- Department of Internal Medicine, Angiology and Physical Medicine, Center for Laser Diagnostics and Therapy, Medical University of Silesia, Bytom, Poland
| | - Aleksandra Kawczyk-Krupka
- Department of Internal Medicine, Angiology and Physical Medicine, Center for Laser Diagnostics and Therapy, Medical University of Silesia, Bytom, Poland
| | - Dorota Bartusik-Aebisher
- Department of Biochemistry and General Chemistry, Medical College of The Rzeszów University, Rzeszów, Poland
| |
Collapse
|
27
|
Wagner S, Ewald C, Freitag D, Herrmann KH, Koch A, Bauer J, Vogl TJ, Kemmling A, Gufler H. Effects of Tetrahydrolipstatin on Glioblastoma in Mice: MRI-Based Morphologic and Texture Analysis Correlated with Histopathology and Immunochemistry Findings-A Pilot Study. Cancers (Basel) 2024; 16:1591. [PMID: 38672673 PMCID: PMC11048907 DOI: 10.3390/cancers16081591] [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: 03/24/2024] [Revised: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND This study aimed to investigate the effects of tetrahydrolipstatin (orlistat) on heterotopic glioblastoma in mice by applying MRI and correlating the results with histopathology and immunochemistry. METHODS Human glioblastoma cells were injected subcutaneously into the groins of immunodeficient mice. After tumor growth of >150 mm3, the animals were assigned into a treatment group (n = 6), which received daily intraperitoneal injections of orlistat, and a control group (n = 7). MRI was performed at the time of randomization and before euthanizing the animals. Tumor volumes were calculated, and signal intensities were analyzed. The internal tumor structure was evaluated visually and with texture analysis. Western blotting and protein expression analysis were performed. RESULTS At histology, all tumors showed high mitotic and proliferative activity (Ki67 ≥ 10%). Reduced fatty acid synthetase expression was measured in the orlistat group (p < 0.05). Based on the results of morphologic MRI-based analysis, tumor growth remained concentric in the control group and changed to eccentric in the treatment group (p < 0.05). The largest area under the receiver operating curve of the predictors derived from the texture analysis of T2w images was for wavelet transform parameters WavEnHL_s3 and WavEnLH_s4 at 0.96 and 1.00, respectively. CONCLUSIONS Orlistat showed effects on heterotopically implanted glioblastoma multiforme in MRI studies of mice based on morphologic and texture analysis.
Collapse
Affiliation(s)
- Sabine Wagner
- Department of Neuroradiology, Marburg University Hospital, Philipps University, 35043 Marburg, Germany;
- Department of Neuroradiology, Institute for Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University, 07747 Jena, Germany
| | - Christian Ewald
- Department of Neurosurgery, Brandenburg Medical School, Campus Brandenburg, 14770 Brandenburg a. d. Havel, Germany (J.B.)
| | - Diana Freitag
- Department of Neurosurgery, Section of Experimental Neurooncology, Jena University Hospital, Friedrich Schiller University, 07747 Jena, Germany;
| | - Karl-Heinz Herrmann
- Medical Physics Group, Institute for Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University, 07743 Jena, Germany;
| | - Arend Koch
- Department of Neuropathology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité University Medicine, 10117 Berlin, Germany
| | - Johannes Bauer
- Department of Neurosurgery, Brandenburg Medical School, Campus Brandenburg, 14770 Brandenburg a. d. Havel, Germany (J.B.)
| | - Thomas J. Vogl
- Department of Diagnostic and Interventional Radiology, Goethe University Hospital Frankfurt, 60590 Frankfurt am Main, Germany; (T.J.V.); (H.G.)
| | - André Kemmling
- Department of Neuroradiology, Marburg University Hospital, Philipps University, 35043 Marburg, Germany;
| | - Hubert Gufler
- Department of Diagnostic and Interventional Radiology, Goethe University Hospital Frankfurt, 60590 Frankfurt am Main, Germany; (T.J.V.); (H.G.)
| |
Collapse
|
28
|
Ishizawa K, Adachi JI, Tamaru JI, Nishikawa R, Mishima K, Sasaki A. Neuropil-like islands are a possible pathogenetic link between glioblastoma and gangliocytoma/ganglioglioma in a case of synchronous bilateral brain tumors. Neuropathology 2024; 44:126-134. [PMID: 37641451 DOI: 10.1111/neup.12939] [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: 04/20/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/31/2023]
Abstract
Neuropil-like islands (NIs) are a histologic hallmark of glioneuronal tumors with neuropil-like islands (GTNIs), but GTNIs are presently not considered a homogeneous entity. The essence of GTNI is likely its glial component, and NIs are now considered aberrant neuronal differentiation or metaplasia. The case we report herein is a 41-year-old woman who was synchronously affected by two brain tumors: one was a glioblastoma (glioblastoma multiforme, GBM), of isocitrate dehydrogenase (IDH)-wild type, with NIs in the left parietal lobe, and the other was histologically a composite gangliocytoma (GC)/anaplastic ganglioglioma (GG) with NIs in the right medial temporal lobe. While both tumors were genetically wild type for IDH, histone H3, and v-raf murine sarcoma viral oncogene homolog B1 (BRAF), the former tumor, but not the latter, was mutated for telomerase reverse transcriptase promoter gene (TERT). A recent systematic study using DNA methylation profiling and next-generation sequencing showed that anaplastic GG separate into other WHO tumor types, including IDH-wild-type GBM. It suggested a diagnostic scheme where an anaplastic GG is likely an IDH-wild-type GBM if it is a BRAF wild type, IDH wild type, and TERT promoter mutant tumor. The likely scenario in this patient is that the GBM results from the progression of GC/anaplastic GG due to the superimposed TERT promoter mutation and the propagation of newly generated GBM cells in the contralateral hemisphere. A systematic analysis using DNA methylation profiling and next-generation sequencing was not available in this study, but the common presence of NIs histologically noted in the two tumors could support this scenario. Although a sufficient volume of molecular and genetic testing is sine qua non for the accurate understanding of brain tumors, the importance of histologic observation cannot be overemphasized.
Collapse
Affiliation(s)
- Keisuke Ishizawa
- Department of Pathology, Saitama Medical University, Moroyama, Japan
| | - Jun-Ichi Adachi
- Department of Neuro-Oncology/Neurosurgery, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Jun-Ichi Tamaru
- Department of Pathology, Saitama Medical Center, Saitama Medical University, Kawagoe, Japan
| | - Ryo Nishikawa
- Department of Neuro-Oncology/Neurosurgery, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Kazuhiko Mishima
- Department of Neuro-Oncology/Neurosurgery, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Atsushi Sasaki
- Department of Pathology, Saitama Medical University, Moroyama, Japan
| |
Collapse
|
29
|
Zhu X, He Y, Wang M, Shu Y, Lai X, Gan C, Liu L. Intratumoral and Peritumoral Multiparametric MRI-Based Radiomics Signature for Preoperative Prediction of Ki-67 Proliferation Status in Glioblastoma: A Two-Center Study. Acad Radiol 2024; 31:1560-1571. [PMID: 37865602 DOI: 10.1016/j.acra.2023.09.010] [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: 06/28/2023] [Revised: 08/28/2023] [Accepted: 09/08/2023] [Indexed: 10/23/2023]
Abstract
RATIONALE AND OBJECTIVES To assess the predictive ability of intratumoral and peritumoral multiparametric magnetic resonance imaging (MRI)-based radiomics signature (RS) for preoperative prediction of Ki-67 proliferation status in glioblastoma. MATERIALS AND METHODS: A total of 205 patients with glioblastoma at two institutions were retrospectively analyzed. Data from institution 1 (n = 158) were used to develop the predictive model, and as an internal test dataset, data from institution 2 (n = 47) constitute the external test dataset. Feature selection was performed using spearman correlation coefficient, univariate ranking method, and the least absolute shrinkage and selection operator algorithm. RSs were established using a logistic regression algorithm. The predictive performance of the RSs was assessed using calibration curve, decision curve analysis (DCA), and area under the curve (AUC). RESULTS In the RSs based on single-parametric (contrast-enhanced T1-weighted image, T2-weighted image, or apparent diffusion coefficient maps), the AUCs of intratumoral, peritumoral, and combined area (intratumoral and peritumoral) were 0.60-0.67, with no significant difference among them. The RSs that using multiparametric features (integrating the previously mentioned three sequences) showed improved AUC compared to the single-parametric RSs; AUC reached 0.75-0.89. Among them, the multiparametric RS based on radiomics features of the combined area (Multi-Com) exhibited the highest performance, with an internal test dataset AUC of 0.89 (95% confidence interval (CI) 0.75-1.00) and an external test dataset AUC of 0.88 (95% CI 0.78-0.97). The calibration curve and DCA display RS (Multi-Com) have good calibration ability and clinical applicability. CONCLUSION The multiparametric MRI-based RS combining intratumoral and peritumoral features can serve as a noninvasive and effective tool for preoperative assessment of Ki-67 proliferation status in glioblastoma.
Collapse
Affiliation(s)
- Xuechao Zhu
- Departments of Radiology, Jiangxi Tumor Hospital, Nanchang, Jiangxi, China (X.Z., Y.S., C.G., L.L.)
| | - Yulin He
- Departments of Radiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (Y.H., M.W., X.L.)
| | - Mengting Wang
- Departments of Radiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (Y.H., M.W., X.L.)
| | - Yuqin Shu
- Departments of Radiology, Jiangxi Tumor Hospital, Nanchang, Jiangxi, China (X.Z., Y.S., C.G., L.L.)
| | - Xunfu Lai
- Departments of Radiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (Y.H., M.W., X.L.)
| | - Cuihua Gan
- Departments of Radiology, Jiangxi Tumor Hospital, Nanchang, Jiangxi, China (X.Z., Y.S., C.G., L.L.)
| | - Lan Liu
- Departments of Radiology, Jiangxi Tumor Hospital, Nanchang, Jiangxi, China (X.Z., Y.S., C.G., L.L.).
| |
Collapse
|
30
|
Lohmeier J, Radbruch H, Brenner W, Hamm B, Hansen B, Tietze A, Makowski MR. Detection of recurrent high-grade glioma using microstructure characteristics of distinct metabolic compartments in a multimodal and integrative 18F-FET PET/fast-DKI approach. Eur Radiol 2024; 34:2487-2499. [PMID: 37672058 PMCID: PMC10957712 DOI: 10.1007/s00330-023-10141-0] [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: 01/29/2023] [Revised: 06/25/2023] [Accepted: 07/06/2023] [Indexed: 09/07/2023]
Abstract
OBJECTIVES Differentiation between high-grade glioma (HGG) and post-treatment-related effects (PTRE) is challenging, but advanced imaging techniques were shown to provide benefit. We aim to investigate microstructure characteristics of metabolic compartments identified from amino acid PET and to evaluate the diagnostic potential of this multimodal and integrative O-(2-18F-fluoroethyl)-L-tyrosine-(FET)-PET and fast diffusion kurtosis imaging (DKI) approach for the detection of recurrence and IDH genotyping. METHODS Fifty-nine participants with neuropathologically confirmed recurrent HGG (n = 39) or PTRE (n = 20) were investigated using static 18F-FET PET and a fast-DKI variant. PET and advanced diffusion metrics of metabolically defined (80-100% and 60-75% areas of 18F-FET uptake) compartments were assessed. Comparative analysis was performed using Mann-Whitney U tests with Holm-Šídák multiple-comparison test and Wilcoxon signed-rank test. Receiver operating characteristic (ROC) curves, regression, and Spearman's correlation analysis were used for statistical evaluations. RESULTS Compared to PTRE, recurrent HGG presented increased 18F-FET uptake and diffusivity (MD60), but lower (relative) mean kurtosis tensor (rMKT60) and fractional anisotropy (FA60) (respectively p < .05). Diffusion metrics determined from the metabolic periphery showed improved diagnostic performance - most pronounced for FA60 (AUC = 0.86, p < .001), which presented similar benefit to 18F-FET PET (AUC = 0.86, p < .001) and was negatively correlated with amino acid uptake (rs = - 0.46, p < .001). When PET and DKI metrics were evaluated in a multimodal biparametric approach, TBRmax + FA60 showed highest diagnostic accuracy (AUC = 0.93, p < .001), which improved the detection of relapse compared to PET alone (difference in AUC = 0.069, p = .04). FA60 and MD60 distinguished the IDH genotype in the post-treatment setting. CONCLUSION Detection of glioma recurrence benefits from a multimodal and integrative PET/DKI approach, which presented significant diagnostic advantage to the assessment based on PET alone. CLINICAL RELEVANCE STATEMENT A multimodal and integrative 18F-FET PET/fast-DKI approach for the non-invasive microstructural characterization of metabolic compartments provided improved diagnostic capability for differentiation between recurrent glioma and post-treatment-related changes, suggesting a role for the diagnostic workup of patients in post-treatment settings. KEY POINTS • Multimodal PET/MRI with integrative analysis of 18F-FET PET and fast-DKI presents clinical benefit for the assessment of CNS cancer, particularly for the detection of recurrent high-grade glioma. • Microstructure markers of the metabolic periphery yielded biologically pertinent estimates characterising the tumour microenvironment, and, thereby, presented improved diagnostic accuracy with similar accuracy to amino acid PET. • Combined 18F-FET PET/fast-DKI achieved the best diagnostic performance for detection of high-grade glioma relapse with significant benefit to the assessment based on PET alone.
Collapse
Affiliation(s)
- Johannes Lohmeier
- Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - Helena Radbruch
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Winfried Brenner
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität Zu Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Bernd Hamm
- Department of Radiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Brian Hansen
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, Universitetsbyen 3, 8000, Aarhus C, Denmark
| | - Anna Tietze
- Institute of Neuroradiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Marcus R Makowski
- Department of Radiology, Technical University Munich, Ismaninger Str. 22, 81675, München, Germany
| |
Collapse
|
31
|
Salvato I, Marchini A. Immunotherapeutic Strategies for the Treatment of Glioblastoma: Current Challenges and Future Perspectives. Cancers (Basel) 2024; 16:1276. [PMID: 38610954 PMCID: PMC11010873 DOI: 10.3390/cancers16071276] [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: 02/28/2024] [Revised: 03/14/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Despite decades of research and the best up-to-date treatments, grade 4 Glioblastoma (GBM) remains uniformly fatal with a patient median overall survival of less than 2 years. Recent advances in immunotherapy have reignited interest in utilizing immunological approaches to fight cancer. However, current immunotherapies have so far not met the anticipated expectations, achieving modest results in their journey from bench to bedside for the treatment of GBM. Understanding the intrinsic features of GBM is of crucial importance for the development of effective antitumoral strategies to improve patient life expectancy and conditions. In this review, we provide a comprehensive overview of the distinctive characteristics of GBM that significantly influence current conventional therapies and immune-based approaches. Moreover, we present an overview of the immunotherapeutic strategies currently undergoing clinical evaluation for GBM treatment, with a specific emphasis on those advancing to phase 3 clinical studies. These encompass immune checkpoint inhibitors, adoptive T cell therapies, vaccination strategies (i.e., RNA-, DNA-, and peptide-based vaccines), and virus-based approaches. Finally, we explore novel innovative strategies and future prospects in the field of immunotherapy for GBM.
Collapse
Affiliation(s)
- Ilaria Salvato
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg;
- Laboratory of Oncolytic Virus Immuno-Therapeutics (LOVIT), Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Antonio Marchini
- Laboratory of Oncolytic Virus Immuno-Therapeutics (LOVIT), Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg
- Laboratory of Oncolytic Virus Immuno-Therapeutics, German Cancer Research Center, 69120 Heidelberg, Germany
| |
Collapse
|
32
|
Ge X, Ma Y, Huang X, Gan T, Ma W, Liu G, Xiong Y, Li M, Wang X, Zhang J. Distinguishment between high-grade gliomas and solitary brain metastases in peritumoural oedema: quantitative analysis using synthetic MRI at 3 T. Clin Radiol 2024; 79:e361-e368. [PMID: 38103981 DOI: 10.1016/j.crad.2023.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 09/12/2023] [Accepted: 10/21/2023] [Indexed: 12/19/2023]
Abstract
AIM To investigate the efficacy of synthetic magnetic resonance imaging (MRI) in distinguishing high-grade gliomas (HGGs) from solitary brain metastases (SBMs) in peritumoural oedema. MATERIALS AND METHODS Thirty-five patients with HGGs and 25 patients with SBMs were recruited and scanned using synthetic MRI using a 3 T scanner. Two radiologists measured synthetic MRI-derived relaxation values independently (T1, T2, proton density [PD]) in the peritumoural oedema, which was used to generate quantitative metrics before (T1native, T2native, and PDnative) and after (T1post, T2post, and PDpost) contrast agent injection. Student's t-test or the Mann-Whitney U-test was performed to detect statistically significant differences in the aforementioned metrics in peritumoural oedema between HGGs and SBMs. The receiver operating characteristic (ROC) curves were plotted to evaluate the efficacy of each metric in distinguishing the two groups, and the areas under the curves (AUCs) were compared pairwise by performing the Delong test. RESULTS The mean T1native, T2native, and T1post values in the peritumoural oedema of HGGs were significantly lower compared with SBMs (all p<0.05). The T1post value had a higher AUC (0.843) in differentiating HGGs and SBMs than all other individual metrics (all p<0.05). The combined T1native, T2native, and T1post model had the best distinguishing performance with an AUC, sensitivity, and specificity of 0.987, 94.3%, and 100%, respectively. CONCLUSIONS Synthetic MRI may be a potential supplement to the preoperative diagnosis of HGGs and SBMs in clinical practice, as the synthetic MRI-derived tri-parametric model in the peritumoural oedema showed significantly improved diagnostic performance in distinguishing HGGs from SBMs.
Collapse
Affiliation(s)
- X Ge
- Second Clinical School, Lanzhou University, Lanzhou 70030, China; Department of Magnetic Resonance, Lanzhou University Second Hospital, Lanzhou 70030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Y Ma
- Department of Magnetic Resonance, Lanzhou University Second Hospital, Lanzhou 70030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - X Huang
- Department of Radiology, General Hospital of Ningxia Medical University, Yinchuan 750003, China
| | - T Gan
- Department of Magnetic Resonance, Lanzhou University Second Hospital, Lanzhou 70030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - W Ma
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, China
| | - G Liu
- Department of Magnetic Resonance, Lanzhou University Second Hospital, Lanzhou 70030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Y Xiong
- GE Healthcare, MR Research, Beijing 100004, China
| | - M Li
- GE Healthcare, MR Enhancement Application, Beijing 100004, China
| | - X Wang
- Department of Radiology, General Hospital of Ningxia Medical University, Yinchuan 750003, China.
| | - J Zhang
- Department of Magnetic Resonance, Lanzhou University Second Hospital, Lanzhou 70030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China.
| |
Collapse
|
33
|
Li Y, Wang J, Song SR, Lv SQ, Qin JH, Yu SC. Models for evaluating glioblastoma invasion along white matter tracts. Trends Biotechnol 2024; 42:293-309. [PMID: 37806896 DOI: 10.1016/j.tibtech.2023.09.005] [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: 05/17/2023] [Revised: 08/31/2023] [Accepted: 09/18/2023] [Indexed: 10/10/2023]
Abstract
White matter tracts (WMs) are one of the main invasion paths of glioblastoma multiforme (GBM). The lack of ideal research models hinders our understanding of the details and mechanisms of GBM invasion along WMs. To date, many potential in vitro models have been reported; nerve fiber culture models and nanomaterial models are biocompatible, and the former have electrically active neurons. Brain slice culture models, organoid models, and microfluidic chip models can simulate the real brain and tumor microenvironment (TME), which contains a variety of cell types. These models are closer to the real in vivo environment and are helpful for further studying not only invasion along WMs by GBM, but also perineural invasion and brain metastasis by solid tumors.
Collapse
Affiliation(s)
- Yao Li
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing 400038, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing 400038, China; Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing 400038, China; Department of Neurosurgery, Xinqiao Hospital, Chongqing 400037, China
| | - Jun Wang
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing 400038, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing 400038, China; Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing 400038, China; Jin-feng Laboratory, Chongqing 401329, China
| | - Si-Rong Song
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing 400038, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing 400038, China; Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing 400038, China
| | - Sheng-Qing Lv
- Department of Neurosurgery, Xinqiao Hospital, Chongqing 400037, China
| | - Jian-Hua Qin
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Niaoning 116023, China.
| | - Shi-Cang Yu
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing 400038, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing 400038, China; Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing 400038, China; Jin-feng Laboratory, Chongqing 401329, China.
| |
Collapse
|
34
|
Bušić M, Rumboldt Z, Čerina D, Bušić Ž, Dolić K. Prognostic Value of Apparent Diffusion Coefficient (ADC) in Patients with Diffuse Gliomas. Cancers (Basel) 2024; 16:681. [PMID: 38398073 PMCID: PMC10886867 DOI: 10.3390/cancers16040681] [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: 01/14/2024] [Revised: 01/30/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
This study aimed to evaluate potential posttreatment changes in ADC values within the tissue surrounding the enhancing lesion, particularly in areas not exhibiting MRI characteristics of involvement. Additionally, the objective was to investigate the correlations among ADC values, treatment response, and survival outcomes in individuals diagnosed with gliomas. This retrospective study included a total of 49 patients that underwent either stereotactic biopsy or maximal surgical resection. Histologically confirmed as Grade III or IV gliomas, all cases adhered to the 2016 and 2021 WHO classifications, with subsequent radio-chemotherapy administered post-surgery. Patients were divided into two groups: short and long survival groups. Baseline and follow-up MRI scans were obtained on a 1.5 T MRI scanner. Two ROI circles were positioned near the enhancing area, one ROI in the NAWM ipsilateral to the neoplasm and another symmetrically in the contralateral hemisphere on ADC maps. At follow-up there was a significant difference in both ipsilateral and contralateral NAWM between the two groups, -0.0857 (p = 0.004) and -0.0607 (p = 0.037), respectively. There was a weak negative correlation between survival and ADC values in ipsilateral and contralateral NAWM at the baseline with the correlation coefficient -0.328 (p = 0.02) and -0.302 (p = 0.04), respectively. The correlation was stronger at the follow-up. The findings indicate that ADC values in normal-appearing white matter (NAWM) may function as a prognostic biomarker in patients with diffuse gliomas.
Collapse
Affiliation(s)
- Marija Bušić
- Department of Diagnostic and Interventional Radiology, University Hospital Split, Spinčićeva 1, 21000 Split, Croatia; (M.B.); (Ž.B.)
| | - Zoran Rumboldt
- School of Medicine, University of Rijeka, Ulica Braće Branchetta 20/1, 51000 Rijeka, Croatia;
| | - Dora Čerina
- Department of Oncology, University Hospital Split, Spinčićeva 1, 21000 Split, Croatia;
| | - Željko Bušić
- Department of Diagnostic and Interventional Radiology, University Hospital Split, Spinčićeva 1, 21000 Split, Croatia; (M.B.); (Ž.B.)
| | - Krešimir Dolić
- Department of Diagnostic and Interventional Radiology, University Hospital Split, Spinčićeva 1, 21000 Split, Croatia; (M.B.); (Ž.B.)
- School of Medicine, University of Split, Šoltanska 1, 21000 Split, Croatia
- University Department of Health Studies, University of Split, Ulica Ruđera Boškovića 35, 21000 Split, Croatia
| |
Collapse
|
35
|
Morales-Gallel R, Ulloa-Navas MJ, García-Tárraga P, Prat-Acín R, Reynés G, Pérez-Borredá P, Rubio L, Capilla-González V, Ferrer-Lozano J, García-Verdugo JM. BCAS1 defines a heterogeneous cell population in diffuse gliomas. Oncotarget 2024; 15:49-64. [PMID: 38275289 PMCID: PMC10812236 DOI: 10.18632/oncotarget.28553] [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: 09/11/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
Oligodendrocyte precursor markers have become of great interest to identify new diagnostic and therapeutic targets for diffuse gliomas, since state-of-the-art studies point towards immature oligodendrocytes as a possible source of gliomagenesis. Brain enriched myelin associated protein 1 (BCAS1) is a novel marker of immature oligodendrocytes and was proposed to contribute to tumorigenesis in non-central nervous system tumors. However, BCAS1 role in diffuse glioma is still underexplored. This study analyzes the expression of BCAS1 in different tumor samples from patients with diffuse gliomas (17 oligodendrogliomas; 8 astrocytomas; 60 glioblastomas) and uncovers the molecular and ultrastructural features of BCAS1+ cells by immunostaining and electron microscopy. Our results show that BCAS1+ cells exhibit stellate or spherical morphology with similar ultrastructural features. Stellate and spherical cells were detected as isolated cells in all studied gliomas. Nevertheless, only stellate cells were found to be proliferative and formed tightly packed nodules with a highly proliferative rate in oligodendrogliomas. Our findings provide a comprehensive characterization of the BCAS1+ cell population within diffuse gliomas. The observed proliferative capacity and distribution of BCAS1+ stellate cells, particularly in oligodendrogliomas, highlight BCAS1 as an interesting marker, warranting further investigation into its role in tumor malignancy.
Collapse
Affiliation(s)
- Raquel Morales-Gallel
- Laboratory of Comparative Neurobiology, Institute Cavanilles of Biodiversity and Evolutionary Biology, University of Valencia-CIBERNED, Valencia, Spain
- These authors contributed equally to this work
| | - María José Ulloa-Navas
- Department of Neurosurgery, Mayo Clinic, Jacksonville, FL 32224, USA
- These authors contributed equally to this work
| | - Patricia García-Tárraga
- Laboratory of Comparative Neurobiology, Institute Cavanilles of Biodiversity and Evolutionary Biology, University of Valencia-CIBERNED, Valencia, Spain
| | - Ricardo Prat-Acín
- Department of Neurosurgery, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Gaspar Reynés
- Group of Clinical and Translational Research in Cancer, Health Research Institute Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Pedro Pérez-Borredá
- Department of Neurosurgery, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Luis Rubio
- Department of Pathology, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Vivian Capilla-González
- Department of Integrative Pathophysiology and Therapies, Andalusian Center for Molecular Biology and Regenerative Medicine-CABIMER, Junta de Andalucía-University of Pablo de Olavide-University of Seville-CSIC, Seville, Spain
| | - Jaime Ferrer-Lozano
- Department of Pathology, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - José Manuel García-Verdugo
- Laboratory of Comparative Neurobiology, Institute Cavanilles of Biodiversity and Evolutionary Biology, University of Valencia-CIBERNED, Valencia, Spain
| |
Collapse
|
36
|
Zheng B, Shao B, Mingrino J, Poggi J, Dowd RS, Anthony DC, Donahue JE, Doberstein CE. BRAF-mutated suprasellar glioblastoma mimicking craniopharyngioma: illustrative case. JOURNAL OF NEUROSURGERY. CASE LESSONS 2024; 7:CASE23649. [PMID: 38252936 PMCID: PMC10805586 DOI: 10.3171/case23649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 12/06/2023] [Indexed: 01/24/2024]
Abstract
BACKGROUND Suprasellar masses commonly include craniopharyngiomas and pituitary adenomas. Suprasellar glioblastoma is exceedingly rare with only a few prior case reports in the literature. Suprasellar glioblastoma can mimic craniopharyngioma or other more common suprasellar etiologies preoperatively. OBSERVATIONS A 65-year-old male with no significant history presented to the emergency department with a subacute decline in mental status. Work-up revealed a large suprasellar mass with extension to the right inferior medial frontal lobe and right lateral ventricle, associated with significant vasogenic edema. The patient underwent an interhemispheric transcallosal approach subtotal resection of the interventricular portion of the mass. Pathological analysis revealed glioblastoma, MGMT partially methylated, with a BRAF V600E mutation. LESSONS Malignant glioblastomas can mimic benign suprasellar masses and should remain on the differential for a diverse set of brain masses with a broad range of radiological and clinical features. For complex cases accessible from the ventricle where the pituitary complex cannot be confidently preserved via a transsphenoidal approach, an interhemispheric approach is also a practical initial surgical option. In addition to providing diagnostic value, molecular profiling may also reveal therapeutically significant gene alterations such as BRAF mutations.
Collapse
Affiliation(s)
- Bryan Zheng
- Department of Neurosurgery, Rhode Island Hospital, Providence, Rhode Island; and
| | - Belinda Shao
- Department of Neurosurgery, Rhode Island Hospital, Providence, Rhode Island; and
| | - Jennifer Mingrino
- Department of Pathology, Rhode Island Hospital, Providence, Rhode Island
| | - Jonathan Poggi
- Department of Neurosurgery, Rhode Island Hospital, Providence, Rhode Island; and
| | - Richard S Dowd
- Department of Neurosurgery, Rhode Island Hospital, Providence, Rhode Island; and
| | - Douglas C Anthony
- Department of Pathology, Rhode Island Hospital, Providence, Rhode Island
| | - John E Donahue
- Department of Pathology, Rhode Island Hospital, Providence, Rhode Island
| | - Curtis E Doberstein
- Department of Neurosurgery, Rhode Island Hospital, Providence, Rhode Island; and
| |
Collapse
|
37
|
Cepeda S. Machine Learning and Radiomics in Gliomas. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1462:231-243. [PMID: 39523269 DOI: 10.1007/978-3-031-64892-2_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The integration of machine learning (ML) and radiomics is emerging as a pivotal advancement in glioma research, offering novel insights into the diagnosis, prognosis, and treatment of these complex tumors. Radiomics involves the extraction of a multitude of quantitative features from medical images. When these features are analyzed through ML algorithms, the precision of tumor characterization is enhanced beyond traditional methods.This chapter examines the application of both supervised and unsupervised ML techniques for interpreting radiomic data, highlighting their potential for accurately predicting tumor grade, identifying genetic mutations, estimating patient survival rates, and evaluating treatment responses. The ability of ML-based radiomic analysis to discern intricate patterns in tumor imaging, imperceptible to human observation, is particularly emphasized.Challenges in this field, including data diversity, overfitting risks, and the need for extensive, annotated datasets, are critically assessed. The necessity of integrating these advanced technologies into clinical practice through interdisciplinary collaboration is underscored as a crucial factor for their effective utilization.Overall, the synergy between ML and radiomics in glioma research represents a significant step toward personalized medicine, offering enhanced tools for patient-specific treatment strategies.
Collapse
Affiliation(s)
- Santiago Cepeda
- Department of Neurosurgery, Río Hortega University Hospital, Valladolid, Spain.
| |
Collapse
|
38
|
van den Elshout R, Ariëns B, Blaauboer J, Meijer FJA, van der Kolk AG, Esmaeili M, Scheenen TWJ, Henssen DJHA. Quantification of perineural satellitosis in pretreatment glioblastoma with structural MRI and a diffusion tensor imaging template. Neurooncol Adv 2024; 6:vdad168. [PMID: 38196738 PMCID: PMC10776201 DOI: 10.1093/noajnl/vdad168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2024] Open
Abstract
Background Survival outcomes for glioblastoma (GBM) patients remain unfavorable, and tumor recurrence is often observed. Understanding the radiological growth patterns of GBM could aid in improving outcomes. This study aimed to examine the relationship between contrast-enhancing tumor growth direction and white matter, using an image registration and deformation strategy. Methods In GBM patients 2 pretreatment scans (diagnostic and neuronavigation) were gathered retrospectively, and coregistered to a template and diffusion tensor imaging (DTI) atlas. The GBM lesions were segmented and coregistered to the same space. Growth vectors were derived and divided into vector populations parallel (Φ = 0-20°) and perpendicular (Φ = 70-90°) to white matter. To test for statistical significance between parallel and perpendicular groups, a paired samples Student's t-test was performed. O6-methylguanine-DNA methyltransferase (MGMT) methylation status and its correlation to growth rate were also tested using a one-way ANOVA test. Results For 78 GBM patients (mean age 61 years ± 13 SD, 32 men), the included GBM lesions showed a predominant preference for perineural satellitosis (P < .001), with a mean percentile growth of 30.8% (95% CI: 29.6-32.0%) parallel (0° < |Φ| < 20°) to white matter. Perpendicular tumor growth with respect to white matter microstructure (70° < |Φ| < 90°) showed to be 22.7% (95% CI: 21.3-24.1%) of total tumor growth direction. Conclusions The presented strategy showed that tumor growth direction in pretreatment GBM patients correlated with white matter architecture. Future studies with patient-specific DTI data are required to verify the accuracy of this method prospectively to identify its usefulness as a clinical metric in pre and posttreatment settings.
Collapse
Affiliation(s)
- Rik van den Elshout
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Benthe Ariëns
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joost Blaauboer
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Frederick J A Meijer
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anja G van der Kolk
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Morteza Esmaeili
- Department of Diagnostic Imaging, Akershus University Hospital, Lørenskog, Norway
- Department of Electrical Engineering and Computer Science, University of Stavanger, Stavanger, Norway
| | - Tom W J Scheenen
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dylan J H A Henssen
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
39
|
Sheikh SF, Akhuj A, Raghuveer R, Saklecha A. Neurophysiotherapy in Grade II Diffuse Astrocytoma: A Case Report. Cureus 2024; 16:e53082. [PMID: 38414688 PMCID: PMC10897357 DOI: 10.7759/cureus.53082] [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: 11/08/2023] [Accepted: 01/27/2024] [Indexed: 02/29/2024] Open
Abstract
Diffuse astrocytoma is a slow, progressive, and invasive tumor that develops from astrocytes and there is no discernible boundary between tumor and brain cells. We present a case of a 48-year-old woman with diffuse astrocytoma who experienced sudden left-sided weakness, multiple convulsive episodes, and vomiting. The patient underwent surgery for a left occipital mini craniotomy with complete tumor removal through a titanium burr hole. Postoperatively, the patient complained of bilateral upper and lower extremities weakness, and decreased muscular tone was found; hence, she was referred to undergo neurophysiotherapy. A four-week rehabilitative protocol was started. Physiotherapy is critical in these patients for ensuring early and rapid recovery and treating the condition's clinical manifestations. The outcome measures employed were the tone grading scale, the Brunnstrom recovery stage, and the Functional Independence Measure (FIM). This case study concludes that physiotherapy rehabilitation for an operated case of grade 2 diffuse astrocytoma led to improved lower limb strength, normal tone, and improved functional independence, which helped the patient achieve better functional activities and a greater quality of life.
Collapse
Affiliation(s)
- Simran F Sheikh
- Department of Neuro-Physiotherapy, Ravi Nair Physiotherapy College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Aditi Akhuj
- Department of Neuro-Physiotherapy, Ravi Nair Physiotherapy College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Raghumahanti Raghuveer
- Department of Neuro-Physiotherapy, Ravi Nair Physiotherapy College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Akshaya Saklecha
- Department of Neuro-Physiotherapy, Ravi Nair Physiotherapy College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
40
|
Ramos-Fresnedo A, Al-Kharboosh R, Twohy EL, Basil AN, Szymkiewicz EC, Zubair AC, Trifiletti DM, Durand N, Dickson DW, Middlebrooks EH, Abarbanel DN, Tzeng SY, Almeida JP, Chaichana KL, Green JJ, Sherman WJ, Quiñones-Hinojosa A. Phase 1, Dose Escalation, Nonrandomized, Open-Label, Clinical Trial Evaluating the Safety and Preliminary Efficacy of Allogenic Adipose-Derived Mesenchymal Stem Cells for Recurrent Glioblastoma: A Clinical Trial Protocol. NEUROSURGERY PRACTICE 2023; 4:e00062. [PMID: 38464470 PMCID: PMC10923529 DOI: 10.1227/neuprac.0000000000000062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 07/18/2023] [Indexed: 02/18/2025]
Abstract
Background and Objectives Despite standard of care with maximal safe resection and chemoradiation, glioblastoma is the most common and aggressive type of primary brain cancer. Surgical resection provides a window of opportunity to locally treat gliomas while the patient is recovering, and before initiating concomitant chemoradiation. To assess the safety and establish the maximum tolerated dose of adipose-derived mesenchymal stem cells (AMSCs) for the treatment of recurrent glioblastoma (GBM). Secondary objectives are to assess the toxicity profile and long-term survival outcomes of patients enrolled in the trial. Additionally, biospecimens will be collected to explore the local and systemic responses to this therapy. Methods We will conduct a phase 1, dose escalated, non-randomized, open label, clinical trial of GBM patients who are undergoing surgical resection for recurrence. Up to 18 patients will receive intra-cavitary application of AMSCs encapsulated in fibrin glue during surgical resection. All patients will be followed for up to 5 years for safety and survival data. Adverse events will be recorded using the CTCAE V5.0. Expected Outcomes This study will explore the maximum tolerated dose (MTD) of AMSCs along with the toxicity profile of this therapy in patients with recurrent GBM. Additionally, preliminary long-term survival and progression-free survival outcome analysis will be used to power further randomized studies. Lastly, CSF and blood will be obtained throughout the treatment period to investigate circulating molecular and inflammatory tumoral/stem cell markers and explore the mechanism of action of the therapeutic intervention. Discussion This prospective translational study will determine the initial safety and toxicity profile of local delivery of AMSCs for recurrent GBM. It will also provide additional survival metrics for future randomized trials.
Collapse
Affiliation(s)
| | | | - Erin L. Twohy
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - Abba C. Zubair
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, Florida, USA
- Center for Regenerative Biotherapeutics, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Nisha Durand
- Center for Regenerative Biotherapeutics, Mayo Clinic, Jacksonville, Florida, USA
| | - Dennis W. Dickson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, Florida, USA
| | - Erik H. Middlebrooks
- Department of Radiology, Neuroradiology Division, Mayo Clinic, Jacksonville, Florida, USA
| | - David N. Abarbanel
- Department of Neurology, Neuro-Oncology Division, Mayo Clinic, Jacksonville, Florida, USA
| | - Stephany Y. Tzeng
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | | - Jordan J. Green
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Wendy J. Sherman
- Department of Neurology, Neuro-Oncology Division, Mayo Clinic, Jacksonville, Florida, USA
| | | |
Collapse
|
41
|
Murnan KM, Horbinski C, Stegh AH. Redox Homeostasis and Beyond: The Role of Wild-Type Isocitrate Dehydrogenases for the Pathogenesis of Glioblastoma. Antioxid Redox Signal 2023; 39:923-941. [PMID: 37132598 PMCID: PMC10654994 DOI: 10.1089/ars.2023.0262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 03/06/2023] [Indexed: 05/04/2023]
Abstract
Significance: Glioblastoma is an aggressive and devastating brain tumor characterized by a dismal prognosis and resistance to therapeutic intervention. To support catabolic processes critical for unabated cellular growth and defend against harmful reactive oxygen species, glioblastoma tumors upregulate the expression of wild-type isocitrate dehydrogenases (IDHs). IDH enzymes catalyze the oxidative decarboxylation of isocitrate to α-ketoglutarate (α-KG), NAD(P)H, and CO2. On molecular levels, IDHs epigenetically control gene expression through effects on α-KG-dependent dioxygenases, maintain redox balance, and promote anaplerosis by providing cells with NADPH and precursor substrates for macromolecular synthesis. Recent Advances: While gain-of-function mutations in IDH1 and IDH2 represent one of the most comprehensively studied mechanisms of IDH pathogenic effects, recent studies identified wild-type IDHs as critical regulators of normal organ physiology and, when transcriptionally induced or down regulated, as contributing to glioblastoma progression. Critical Issues: Here, we will discuss molecular mechanisms of how wild-type IDHs control glioma pathogenesis, including the regulation of oxidative stress and de novo lipid biosynthesis, and provide an overview of current and future research directives that aim to fully characterize wild-type IDH-driven metabolic reprogramming and its contribution to the pathogenesis of glioblastoma. Future Directions: Future studies are required to further dissect mechanisms of metabolic and epigenomic reprogramming in tumors and the tumor microenvironment, and to develop pharmacological approaches to inhibit wild-type IDH function. Antioxid. Redox Signal. 39, 923-941.
Collapse
Affiliation(s)
- Kevin M. Murnan
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Malnati Brain Tumor Institute, Northwestern University, Chicago, Illinois, USA
| | - Craig Horbinski
- Department of Pathology, Feinberg School of Medicine, Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Feinberg School of Medicine, Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Alexander H. Stegh
- Department of Neurological Surgery, The Brain Tumor Center, Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
42
|
Hoshimaru T, Nonoguchi N, Kosaka T, Furuse M, Kawabata S, Yagi R, Kurisu Y, Kashiwagi H, Kameda M, Takami T, Kataoka-Sasaki Y, Sasaki M, Honmou O, Hiramatsu R, Wanibuchi M. Actin Alpha 2, Smooth Muscle (ACTA2) Is Involved in the Migratory Potential of Malignant Gliomas, and Its Increased Expression at Recurrence Is a Significant Adverse Prognostic Factor. Brain Sci 2023; 13:1477. [PMID: 37891844 PMCID: PMC10605410 DOI: 10.3390/brainsci13101477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/17/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Malignant glioma is a highly invasive tumor, and elucidating the glioma invasion mechanism is essential for developing novel therapies. We aimed to highlight actin alpha 2, smooth muscle (ACTA2) as potential biomarkers of brain invasion and distant recurrence in malignant gliomas. Using the human malignant glioma cell line, U251MG, we generated ACTA2 knockdown (KD) cells treated with small interfering RNA, and the cell motility and proliferation of the ACTA2 KD group were analyzed. Furthermore, tumor samples from 12 glioma patients who underwent reoperation at the time of tumor recurrence were utilized to measure ACTA2 expression in the tumors before and after recurrence. Thereafter, we examined how ACTA2 expression correlates with the time to tumor recurrence and the mode of recurrence. The results showed that the ACTA2 KD group demonstrated a decline in the mean motion distance and proliferative capacity compared to the control group. In the clinical glioma samples, ACTA2 expression was remarkably increased in recurrent samples compared to the primary samples from the same patients, and the higher the change in ACTCA2 expression from the start to relapse, the shorter the progression-free survival. In conclusion, ACTA2 may be involved in distant recurrence in clinical gliomas.
Collapse
Affiliation(s)
- Takumi Hoshimaru
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| | - Naosuke Nonoguchi
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| | - Takuya Kosaka
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| | - Motomasa Furuse
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| | - Shinji Kawabata
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| | - Ryokichi Yagi
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| | - Yoshitaka Kurisu
- Department of Pathology, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| | - Hideki Kashiwagi
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| | - Masahiro Kameda
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| | - Toshihiro Takami
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| | - Yuko Kataoka-Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Hokkaido 060-8556, Japan
| | - Masanori Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Hokkaido 060-8556, Japan
| | - Osamu Honmou
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Hokkaido 060-8556, Japan
| | - Ryo Hiramatsu
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| | - Masahiko Wanibuchi
- Department of Neurosurgery, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan
| |
Collapse
|
43
|
Zahoor W, Pandith AA, Nisar S, Qasim I, Surana M, Ganie FA, Manzoor U, Arif SH, Rasool SUA, Lateef A, Shah P, Bhat RA. EPIDERMAL GROWTH FACTOR RECEPTOR (EGFR) POLYMORPHIC VARIATIONS (-216G/T & -191 C/A) POSE A HIGH RISK TO PATIENTS WITH MALIGNANT GLIOMA. Exp Oncol 2023; 45:203-210. [PMID: 37824771 DOI: 10.15407/exp-oncology.2023.02.203] [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: 10/10/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND Malignant gliomas are the most frequent and lethal brain tumors. Their molecular aspects remain intangible but current studies have pointed to certain genetic polymorphic loci that pose the risk. The polymorphic sequence variations of the epidermal growth factor receptor gene (EGFR) pathway play a vital role in the glioma risk, and the EGFR variants (216G>T and 191C>A) are identified to affect the risk for the development of different tumors including glioma. AIM To examine genetic variations of EGFR T rs712829 (216G/T) and rs712830 (191C>A) with respect to glioma risk. MATERIALS AND METHODS 129 confirmed glioma cases were genotyped against 180 malignancy-free healthy controls by polymerase chain reaction-restriction fragment length polymorphism technique (RFLP). RESULTS The frequency of the TT homozygous variant of the EGFR -216 G/T genotype differed significantly between cases and controls (49.6% vs. 23.0%) (p < 0.0001). The EGFR -216 G>T allele 'T' was found significantly more frequently in cases (0.56 vs. 0.33 in controls; p < 0.0001). The EGFR -191C>A homozygous 'AA' genotype was implicated significantly more frequently in cases than in controls (p < 0.0001). The distribution of the 'A' variant allele was also more frequent in cases (41.9%) than in controls (14.0%) (0.55 vs. 0.30; p < 0.0001). TC and TA haplotypes showed varied frequency in cases and controls. CONCLUSION EGFR -216 G>T and -191 C>A variants and haplotypes (TA and TC) of the EGFR gene are very strong risk factors in the development of glioma in the Kashmiri population.
Collapse
Affiliation(s)
- Wani Zahoor
- Advanced Centre for Human Genetics, Sher-I-Kashmir Institute of Medical Sciences, Srinagar 190011, J&K, India
- Department of Chemistry, Mewar University-Jaipur, India
| | - Arshad A Pandith
- Advanced Centre for Human Genetics, Sher-I-Kashmir Institute of Medical Sciences, Srinagar 190011, J&K, India
| | - Syed Nisar
- Medical Oncology, Sher-I-Kashmir Institute of Medical Sciences, Srinagar 190011, J&K, India
| | - Iqbal Qasim
- Advanced Centre for Human Genetics, Sher-I-Kashmir Institute of Medical Sciences, Srinagar 190011, J&K, India
| | - Menka Surana
- Department of Chemistry, Mewar University-Jaipur, India
| | - Farooq A Ganie
- Department of CVTS, Sher-I-Kashmir Institute of Medical Sciences, Srinagar 190011, J&K, India
| | - Usma Manzoor
- Advanced Centre for Human Genetics, Sher-I-Kashmir Institute of Medical Sciences, Srinagar 190011, J&K, India
| | - Sajad H Arif
- Department of Neurosurgery, Sher-I-Kashmir Institute of Medical Sciences, Srinagar 190011, J&K, India
| | - Shayaq Ul Abeer Rasool
- Advanced Centre for Human Genetics, Sher-I-Kashmir Institute of Medical Sciences, Srinagar 190011, J&K, India
| | - Adil Lateef
- Advanced Centre for Human Genetics, Sher-I-Kashmir Institute of Medical Sciences, Srinagar 190011, J&K, India
| | - Parveen Shah
- Department of Pathology, Sher-I-Kashmir Institute of Medical Sciences, Srinagar 190011, J&K, India
| | - Rashid A Bhat
- Department of Neurosurgery, Sher-I-Kashmir Institute of Medical Sciences, Srinagar 190011, J&K, India
| |
Collapse
|
44
|
Amjad G, Zeinali Zadeh M, Azmoudeh-Ardalan F, Jalali AH, Shakiba M, Ghavami N, Oghabian Z, Oghabian MA, Firouznia S, Rafiei B, Sabet Rasekh P, Tahmasebi Arashloo F, Firouznia K. Evaluation of multimodal MR imaging for differentiating infiltrative versus reactive edema in brain gliomas. Br J Neurosurg 2023; 37:1031-1039. [PMID: 33263433 DOI: 10.1080/02688697.2020.1849541] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 11/05/2020] [Indexed: 01/28/2023]
Abstract
OBJECTIVE To determine the border of glial tumors by diffusion weighted imaging (DWI), apparent diffusion co-efficient (ADC), magnetic resonance spectroscopy (MRS) and perfusion brain MRI. PATIENTS AND METHODS Ten patients with brain gliomas were enrolled [mean age: 35.3 ± 13.2, range: 20-62]. Conventional MRI was performed for all patients. Besides, tumor mapping based on Choline (Cho)/Creatine (Cr) color map in MRS, perfusion and diffusion color maps, were gathered. Different tumoral and peritumoral regions [normal tissue, reactive edema, infiltrative edema, and tumor core] were defined. MRI criteria were evaluated in areas targeted for biopsy and histopathologic evaluation was determined. RESULTS Tumor cell positive samples [one necrosis, 26 infiltrative and nine tumor cores] composed 36 (75%) of the 48 samples. Seven (19.4%) of the positive samples were interpreted as not tumor on MRI. Five were identified as reactive edema and two as normal tissue] [kappa: .67, p-value < .001]. Mean of ADC, median of N-acetylaspartate (NAA) and NAA/Cho were statistically different between positive and negative samples (p = .02 and p < .001, respectively). Mean ADC and median Cho/NAA were statistically different in missed tumor containing tissue presented as reactive edema compared to normal and correctly diagnosed reactive edema samples together (p-values < .05). CONCLUSIONS Multimodal MRI could define infiltrated borders of brain gliomas.
Collapse
Affiliation(s)
- Ghazaleh Amjad
- Shahid Akbar Abadi Clinical Research Development Unit (ShCRDU), Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mehdi Zeinali Zadeh
- Department of Neurosurgery, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Azmoudeh-Ardalan
- Department of Pathology, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Jalali
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Madjid Shakiba
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Nafiseh Ghavami
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Zeynab Oghabian
- Neuroimaging and Analysis Group Research Center, Molecular and Cellular Imaging Department, Medical Physics and Biomedical Engineering Department, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Oghabian
- Neuroimaging and Analysis Group Research Center, Molecular and Cellular Imaging Department, Medical Physics and Biomedical Engineering Department, Tehran University of Medical Sciences, Tehran, Iran
| | - Saba Firouznia
- Department of Engineering Mathematics, University of Bristol, Bristol, UK
| | - Behrouz Rafiei
- Medical Physics, Tehran University of Medical Sciences, Tehran, Iran
| | - Parto Sabet Rasekh
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Kavous Firouznia
- Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
45
|
McDonald B, Barth K, Schmidt MHH. The origin of brain malignancies at the blood-brain barrier. Cell Mol Life Sci 2023; 80:282. [PMID: 37688612 PMCID: PMC10492883 DOI: 10.1007/s00018-023-04934-1] [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: 05/17/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/11/2023]
Abstract
Despite improvements in extracranial therapy, survival rate for patients suffering from brain metastases remains very poor. This is coupled with the incidence of brain metastases continuing to rise. In this review, we focus on core contributions of the blood-brain barrier to the origin of brain metastases. We first provide an overview of the structure and function of the blood-brain barrier under physiological conditions. Next, we discuss the emerging idea of a pre-metastatic niche, namely that secreted factors and extracellular vesicles from a primary tumor site are able to travel through the circulation and prime the neurovasculature for metastatic invasion. We then consider the neurotropic mechanisms that circulating tumor cells possess or develop that facilitate disruption of the blood-brain barrier and survival in the brain's parenchyma. Finally, we compare and contrast brain metastases at the blood-brain barrier to the primary brain tumor, glioma, examining the process of vessel co-option that favors the survival and outgrowth of brain malignancies.
Collapse
Affiliation(s)
- Brennan McDonald
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Dresden, Germany.
| | - Kathrin Barth
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Dresden, Germany
| | - Mirko H H Schmidt
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Dresden, Germany
| |
Collapse
|
46
|
Bartusik-Aebisher D, Woźnicki P, Dynarowicz K, Aebisher D. Photosensitizers for Photodynamic Therapy of Brain Cancers-A Review. Brain Sci 2023; 13:1299. [PMID: 37759900 PMCID: PMC10526171 DOI: 10.3390/brainsci13091299] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
On average, there are about 300,000 new cases of brain cancer each year. Studies have shown that brain and central nervous system tumors are among the top ten causes of death. Due to the extent of this problem and the percentage of patients suffering from brain tumors, innovative therapeutic treatment methods are constantly being sought. One such innovative therapeutic method is photodynamic therapy (PDT). Photodynamic therapy is an alternative and unique technique widely used in dermatology and other fields of medicine for the treatment of oncological and nononcological lesions. Photodynamic therapy consists of the destruction of cancer cells and inducing inflammatory changes by using laser light of a specific wavelength in combination with the application of a photosensitizer. The most commonly used photosensitizers include 5-aminolevulinic acid for the enzymatic generation of protoporphyrin IX, Temoporfin-THPC, Photofrin, Hypericin and Talaporfin. This paper reviews the photosensitizers commonly used in photodynamic therapy for brain tumors. An overview of all three generations of photosensitizers is presented. Along with an indication of the limitations of the treatment of brain tumors, intraoperative photodynamic therapy and its possibilities are described as an alternative therapeutic method.
Collapse
Affiliation(s)
- Dorota Bartusik-Aebisher
- Department of Biochemistry and General Chemistry, Medical College of the University of Rzeszów, 35-959 Rzeszów, Poland;
| | - Paweł Woźnicki
- Students English Division Science Club, Medical College of the University of Rzeszów, 35-959 Rzeszów, Poland;
| | - Klaudia Dynarowicz
- Center for Innovative Research in Medical and Natural Sciences, Medical College of the University of Rzeszów, 35-310 Rzeszów, Poland;
| | - David Aebisher
- Department of Photomedicine and Physical Chemistry, Medical College of the University of Rzeszów, 35-959 Rzeszów, Poland
| |
Collapse
|
47
|
Michelucci A, Sforna L, Di Battista A, Franciolini F, Catacuzzeno L. Ca 2+ -activated K + channels regulate cell volume in human glioblastoma cells. J Cell Physiol 2023; 238:2120-2134. [PMID: 37431808 DOI: 10.1002/jcp.31072] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/10/2023] [Accepted: 06/20/2023] [Indexed: 07/12/2023]
Abstract
Glioblastoma (GBM), the most lethal form of brain tumors, bases its malignancy on the strong ability of its cells to migrate and invade the narrow spaces of healthy brain parenchyma. Cell migration and invasion are both critically dependent on changes in cell volume and shape driven by the transmembrane transport of osmotically important ions such as K+ and Cl- . However, while the Cl- channels participating in cell volume regulation have been clearly identified, the precise nature of the K+ channels involved is still uncertain. Using a combination of electrophysiological and imaging approaches in GBM U87-MG cells, we found that hypotonic-induced cell swelling triggered the opening of Ca2+ -activated K+ (KCa ) channels of large and intermediate conductance (BKCa and IKCa , respectively), both highly expressed in GBM cells. The influx of Ca2+ mediated by the hypotonic-induced activation of mechanosensitive channels was found to be a key step for opening both the BKCa and the IKCa channels. Finally, the activation of both KCa channels mediated by mechanosensitive channels was found to be essential for the development of the regulatory volume decrease following hypotonic shock. Taken together, these data indicate that KCa channels are the main K+ channels responsible for the volume regulation in U87-MG cells.
Collapse
Affiliation(s)
- Antonio Michelucci
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Perugia, Italy
| | - Luigi Sforna
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Perugia, Italy
| | - Angela Di Battista
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Perugia, Italy
| | - Fabio Franciolini
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Perugia, Italy
| | - Luigi Catacuzzeno
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Perugia, Italy
| |
Collapse
|
48
|
Vadla R, Miki S, Taylor B, Kawauchi D, Jones BM, Nathwani N, Pham P, Tsang J, Nathanson DA, Furnari FB. Glioblastoma Mesenchymal Transition and Invasion are Dependent on a NF-κB/BRD2 Chromatin Complex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.03.546613. [PMID: 37461511 PMCID: PMC10349949 DOI: 10.1101/2023.07.03.546613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Glioblastoma (GBM) represents the most aggressive subtype of glioma, noted for its profound invasiveness and molecular heterogeneity. The mesenchymal (MES) transcriptomic subtype is frequently associated with therapy resistance, rapid recurrence, and increased tumor-associated macrophages. Notably, activation of the NF-κB pathway and alterations in the PTEN gene are both associated with this malignant transition. Although PTEN aberrations have been shown to be associated with enhanced NF-κB signaling, the relationships between PTEN, NF-κB and MES transition are poorly understood in GBM. Here, we show that PTEN regulates the chromatin binding of bromodomain and extraterminal (BET) family proteins, BRD2 and BRD4, mediated by p65/RelA localization to the chromatin. By utilizing patient-derived glioblastoma stem cells and CRISPR gene editing of the RELA gene, we demonstrate a crucial role for RelA lysine 310 acetylation in recruiting BET proteins to chromatin for MES gene expression and GBM cell invasion upon PTEN loss. Remarkably, we found that BRD2 is dependent on chromatin associated acetylated RelA for its recruitment to MES gene promoters and their expression. Furthermore, loss of BRD2 results in the loss of MES signature, accompanied by an enrichment of proneural signature and enhanced therapy responsiveness. Finally, we demonstrate that disrupting the NFκB/BRD2 interaction with a brain penetrant BET-BD2 inhibitor reduces mesenchymal gene expression, GBM invasion, and therapy resistance in GBM models. This study uncovers the role of hitherto unexplored PTEN-NF-κB-BRD2 pathway in promoting MES transition and suggests inhibiting this complex with BET-BD2 specific inhibitors as a therapeutic approach to target the MES phenotype in GBM.
Collapse
Affiliation(s)
- Raghavendra Vadla
- Division of Regenerative Medicine, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Shunichiro Miki
- Division of Regenerative Medicine, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Brett Taylor
- Division of Regenerative Medicine, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Daisuke Kawauchi
- Division of Regenerative Medicine, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Brandon M Jones
- Division of Regenerative Medicine, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Nidhi Nathwani
- Division of Regenerative Medicine, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Philip Pham
- Division of Regenerative Medicine, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jonathan Tsang
- Departments of Molecular and Medical Pharmacology, University of California, Los Angeles, California 90095, United States
| | - David A Nathanson
- Departments of Molecular and Medical Pharmacology, University of California, Los Angeles, California 90095, United States
| | - Frank B Furnari
- Division of Regenerative Medicine, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
49
|
Hu D, Xia M, Wu L, Liu H, Chen Z, Xu H, He C, Wen J, Xu X. Challenges and advances for glioma therapy based on inorganic nanoparticles. Mater Today Bio 2023; 20:100673. [PMID: 37441136 PMCID: PMC10333687 DOI: 10.1016/j.mtbio.2023.100673] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/10/2023] [Accepted: 05/18/2023] [Indexed: 07/15/2023] Open
Abstract
Glioma is one of the most serious central nervous system diseases, with high mortality and poor prognosis. Despite the continuous development of existing treatment methods, the median survival time of glioma patients is still only 15 months. The main treatment difficulties are the invasive growth of glioma and the obstruction of the blood-brain barrier (BBB) to drugs. With rapid advancements in nanotechnology, inorganic nanoparticles (INPs) have shown favourable application prospects in the diagnosis and treatment of glioma. Due to their extraordinary intrinsic features, INPs can be easily fabricated, while doping with other elements and surface modification by biological ligands can be used to enhance BBB penetration, targeted delivery and biocompatibility. Guided glioma theranostics with INPs can improve and enhance the efficacy of traditional methods such as chemotherapy, radiotherapy and gene therapy. New strategies, such as immunotherapy, photothermal and photodynamic therapy, magnetic hyperthermia therapy, and multifunctional inorganic nanoplatforms, have also been facilitated by INPs. This review emphasizes the current state of research and clinical applications of INPs, including glioma targeting and BBB penetration enhancement methods, in vivo and in vitro biocompatibility, and diagnostic and treatment strategies. As such, it provides insights for the development of novel glioma treatment strategies.
Collapse
Affiliation(s)
- Die Hu
- Key Laboratory of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Miao Xia
- Key Laboratory of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Linxuan Wu
- Key Laboratory of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Hanmeng Liu
- Key Laboratory of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Zhigang Chen
- Key Laboratory of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Hefeng Xu
- Key Laboratory of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, 110122, China
| | - Chuan He
- Department of Laboratory Medicine, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Jian Wen
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Xiaoqian Xu
- Key Laboratory of Cell Biology, Ministry of Public Health and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, 110122, China
| |
Collapse
|
50
|
Miyai M, Iwama T, Hara A, Tomita H. Exploring the Vital Link Between Glioma, Neuron, and Neural Activity in the Context of Invasion. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:669-679. [PMID: 37286277 DOI: 10.1016/j.ajpath.2023.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/15/2023] [Accepted: 02/23/2023] [Indexed: 06/09/2023]
Abstract
Because of their ability to infiltrate normal brain tissue, gliomas frequently evade microscopic surgical excision. The histologic infiltrative property of human glioma has been previously characterized as Scherer secondary structures, of which the perivascular satellitosis is a prospective target for anti-angiogenic treatment in high-grade gliomas. However, the mechanisms underlying perineuronal satellitosis remain unclear, and therapy remains lacking. Our knowledge of the mechanism underlying Scherer secondary structures has improved over time. New techniques, such as laser capture microdissection and optogenetic stimulation, have advanced our understanding of glioma invasion mechanisms. Although laser capture microdissection is a useful tool for studying gliomas that infiltrate the normal brain microenvironment, optogenetics and mouse xenograft glioma models have been extensively used in studies demonstrating the unique role of synaptogenesis in glioma proliferation and identification of potential therapeutic targets. Moreover, a rare glioma cell line is established that, when transplanted in the mouse brain, can replicate and recapitulate the human diffuse invasion phenotype. This review discusses the primary molecular causes of glioma, its histopathology-based invasive mechanisms, and the importance of neuronal activity and interactions between glioma cells and neurons in the brain microenvironment. It also explores current methods and models of gliomas.
Collapse
Affiliation(s)
- Masafumi Miyai
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan; Department of Neurosurgery, Hashima City Hospital, Gifu, Japan; Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Toru Iwama
- Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Akira Hara
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan.
| |
Collapse
|