Neuro-oncology advancesPub Date : 2025-01-29eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf025
Sophie Shih-Yüng Wang, Gerhard Horstmann, Mirjam Renovanz, Albertus van Eck, Marcos Tatagiba, Georgios Naros
{"title":"Sex-specific difference in treatment success/failure after vestibular schwannoma treatment.","authors":"Sophie Shih-Yüng Wang, Gerhard Horstmann, Mirjam Renovanz, Albertus van Eck, Marcos Tatagiba, Georgios Naros","doi":"10.1093/noajnl/vdaf025","DOIUrl":"https://doi.org/10.1093/noajnl/vdaf025","url":null,"abstract":"<p><strong>Background: </strong>Sex-related differences in patients with sporadic, unilateral vestibular schwannoma (VS) are poorly investigated so far, and it remains unclear whether biological sex affects treatment response to stereotactic radiosurgery (<i>SRS</i>) or microsurgical resection (<i>SURGERY</i>). This study elucidates sex-related differences in treatment outcome of VS.</p><p><strong>Methods: </strong>This is a retrospective two-center cohort study. All consecutive patients treated for their VS between 2005 and 2012 were included. Previously treated VS and patients with neurofibromatosis were excluded. Clinical status and treatment-related complications were analyzed from both centers' prospective treatment registries. Recurrence/progression-free-survival was assessed radiographically by contrast-enhanced magnetic resonance imaging.</p><p><strong>Results: </strong>Within the entire patient cohort of <i>N</i> = 1,118, the majority of VS patients (56%) was female. Sixty-two percent of patients were treated by <i>SRS</i>. Females with very small tumors (KOOS I) were significantly less likely to be assigned to <i>SURGERY</i> than males (<i>P = </i>.009). Mean follow-up time was 6 ± 4.3 years. In <i>SURGERY</i>, the rate of subtotal resection was significantly higher in women (7%) compared to men (2%) (<i>P = </i>.041). However, there was no difference in long-term tumor control after <i>SURGERY</i> between both sexes (<i>P = </i>.729). In <i>SRS</i> however, the incidence of recurrence was significantly higher in women (14%) compared to men (8%) (<i>P</i> = .004), which was also reflected in the Kaplan-Meier analysis (<i>P</i> = .031).</p><p><strong>Conclusions: </strong>Female sex was a negative prognostic factor for treatment success (long-term tumor control) if treated with <i>SRS</i>-there was no sex-related differences in long-term tumor control after <i>SURGERY</i>. Additional research is needed to elucidate sex-related differences in tumor biology affecting the response to VS treatment.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf025"},"PeriodicalIF":3.7,"publicationDate":"2025-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12035609/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144033936","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Neuro-oncology advancesPub Date : 2025-01-29eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf024
Eduardo Rodriguez Almaraz, Geno A Guerra, Nadeem N Al-Adli, Jacob S Young, Abraham Dada, Daniel Quintana, Jennie W Taylor, Nancy Ann Oberheim Bush, Jennifer L Clarke, Nicholas A Butowski, John de Groot, Melike Pekmezci, Arie Perry, Andrew W Bollen, Aaron W Scheffler, David V Glidden, Joanna J Phillips, Joseph F Costello, Edward F Chang, Shawn Hervey-Jumper, Mitchel S Berger, Stephen S Francis, Susan M Chang, David A Solomon
{"title":"Longitudinal profiling of IDH-mutant astrocytomas reveals acquired RAS-MAPK pathway mutations associated with inferior survival.","authors":"Eduardo Rodriguez Almaraz, Geno A Guerra, Nadeem N Al-Adli, Jacob S Young, Abraham Dada, Daniel Quintana, Jennie W Taylor, Nancy Ann Oberheim Bush, Jennifer L Clarke, Nicholas A Butowski, John de Groot, Melike Pekmezci, Arie Perry, Andrew W Bollen, Aaron W Scheffler, David V Glidden, Joanna J Phillips, Joseph F Costello, Edward F Chang, Shawn Hervey-Jumper, Mitchel S Berger, Stephen S Francis, Susan M Chang, David A Solomon","doi":"10.1093/noajnl/vdaf024","DOIUrl":"10.1093/noajnl/vdaf024","url":null,"abstract":"<p><strong>Background: </strong>Isocitrate dehydrogenase (IDH)-mutant astrocytomas represent the most frequent primary intraparenchymal brain tumor in young adults, which typically arise as low-grade neoplasms that often progress and transform to higher grade despite current therapeutic approaches. However, the genetic alterations underlying high-grade transformation and disease progression of IDH-mutant astrocytomas remain inadequately defined.</p><p><strong>Methods: </strong>Genomic profiling was performed on 205 IDH-mutant astrocytomas from 172 patients from both initial treatment-naive and recurrent post-treatment tumor specimens. Molecular findings were integrated with clinical outcomes and pathologic features to define the associations of novel genetic alterations in the RAS-MAPK signaling pathway.</p><p><strong>Results: </strong>Likely oncogenic alterations within the RAS-MAPK mitogenic signaling pathway were identified in 13% of IDH-mutant astrocytomas, which involved the <i>KRAS, NRAS, BRAF, NF1</i>, <i>SPRED1</i>, and <i>LZTR1</i> genes. These included focal amplifications and known activating mutations in oncogenic components (e.g. <i>KRAS</i>, <i>BRAF</i>), as well as deletions and truncating mutations in negative regulatory components (e.g. <i>NF1</i>, <i>SPRED1</i>). These RAS-MAPK pathway alterations were enriched in recurrent tumors and occurred nearly always in high-grade tumors, often co-occurring with <i>CDKN2A</i> homozygous deletion. Patients whose IDH-mutant astrocytomas harbored these oncogenic RAS-MAPK pathway alterations had inferior survival compared to those with RAS-MAPK wild-type tumors.</p><p><strong>Conclusions: </strong>These findings highlight novel genetic perturbations in the RAS-MAPK pathway as a likely mechanism contributing to the high-grade transformation and treatment resistance of IDH-mutant astrocytomas that may be a potential therapeutic target for affected patients and used for future risk stratification.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf024"},"PeriodicalIF":3.7,"publicationDate":"2025-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11883348/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143575061","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Neuro-oncology advancesPub Date : 2025-01-28eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdae197
Meryem Abbad Andaloussi, Raphael Maser, Frank Hertel, François Lamoline, Andreas Dominik Husch
{"title":"Exploring adult glioma through MRI: A review of publicly available datasets to guide efficient image analysis.","authors":"Meryem Abbad Andaloussi, Raphael Maser, Frank Hertel, François Lamoline, Andreas Dominik Husch","doi":"10.1093/noajnl/vdae197","DOIUrl":"10.1093/noajnl/vdae197","url":null,"abstract":"<p><strong>Background: </strong>Publicly available data are essential for the progress of medical image analysis, in particular for crafting machine learning models. Glioma is the most common group of primary brain tumors, and magnetic resonance imaging (MRI) is a widely used modality in their diagnosis and treatment. However, the availability and quality of public datasets for glioma MRI are not well known.</p><p><strong>Methods: </strong>In this review, we searched for public datasets of glioma MRI using Google Dataset Search, The Cancer Imaging Archive, and Synapse.</p><p><strong>Results: </strong>A total of 28 datasets published between 2005 and May 2024 were found, containing 62 019 images from 5515 patients. We analyzed the characteristics of these datasets, such as the origin, size, format, annotation, and accessibility. Additionally, we examined the distribution of tumor types, grades, and stages among the datasets. The implications of the evolution of the World Health Organization (WHO) classification on tumors of the brain are discussed, in particular the 2021 update that significantly changed the definition of glioblastoma.</p><p><strong>Conclusions: </strong>Potential research questions that could be explored using these datasets were highlighted, such as tumor evolution through malignant transformation, MRI normalization, and tumor segmentation. Interestingly, only 2 datasets among the 28 studied reflect the current WHO classification. This review provides a comprehensive overview of the publicly available datasets for glioma MRI currently at our disposal, providing aid to medical image analysis researchers in their decision-making on efficient dataset choice.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdae197"},"PeriodicalIF":3.7,"publicationDate":"2025-01-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11773385/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143061789","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Neuro-oncology advancesPub Date : 2025-01-27eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf010
Grzegorz Wicher, Ananya Roy, Alessandra Vaccaro, Kalyani Vemuri, Mohanraj Ramachandran, Tommie Olofsson, Rebeca-Noemi Imbria, Mattias Belting, Gunnar Nilsson, Anna Dimberg, Karin Forsberg-Nilsson
{"title":"Lack of ST2 aggravates glioma invasiveness, vascular abnormality, and immune suppression.","authors":"Grzegorz Wicher, Ananya Roy, Alessandra Vaccaro, Kalyani Vemuri, Mohanraj Ramachandran, Tommie Olofsson, Rebeca-Noemi Imbria, Mattias Belting, Gunnar Nilsson, Anna Dimberg, Karin Forsberg-Nilsson","doi":"10.1093/noajnl/vdaf010","DOIUrl":"10.1093/noajnl/vdaf010","url":null,"abstract":"<p><strong>Background: </strong>Glioblastoma (GBM) is the most common primary malignant brain tumor in adults, characterized by aggressive growth and a dismal prognosis. Interleukin-33 (IL-33) and its receptor ST2 have emerged as regulators of glioma growth, but their exact function in tumorigenesis has not been deciphered. Indeed, previous studies on IL-33 in cancer have yielded somewhat opposing results as to whether it is pro- or anti-tumorigenic.</p><p><strong>Methods: </strong>IL-33 expression was assessed in a GBM tissue microarray and public databases. As in vivo models we used orthotopic xenografts of patient-derived GBM cells, and syngenic models with grafted mouse glioma cells.</p><p><strong>Results: </strong>We analyzed the role of IL-33 and its receptor ST2 in nonmalignant cells of the glioma microenvironment and found that IL-33 levels are increased in cells surrounding the tumor. Protein complexes of IL-33 and ST2 are mainly found outside of the tumor core. The IL-33-producing cells consist primarily of oligodendrocytes. To determine the function of IL-33 in the tumor microenvironment, we used mice lacking the ST2 receptor. When glioma cells were grafted to ST2-deficient mouse brains, the resulting tumors exhibited a more invasive growth pattern, and are associated with poorer survival, compared to <i>wild-type</i> mice. Tumors in ST2-deficient hosts are more invasive, with increased expression of extracellular matrix remodeling enzymes and enhanced tumor angiogenesis. Furthermore, the absence of ST2 leads to a more immunosuppressive environment.</p><p><strong>Conclusions: </strong>Our findings reveal that glia-derived IL-33 and its receptor ST2 participate in modulating tumor invasiveness, tumor vasculature, and immunosuppression in glioma.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf010"},"PeriodicalIF":3.7,"publicationDate":"2025-01-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11808570/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143392464","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Neuro-oncology advancesPub Date : 2025-01-25eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf017
Omar Rafael Alemán, Juan Carlos Quintero, Ignacio Camacho-Arroyo
{"title":"The language of glioblastoma: A tale of cytokines and sex hormones communication.","authors":"Omar Rafael Alemán, Juan Carlos Quintero, Ignacio Camacho-Arroyo","doi":"10.1093/noajnl/vdaf017","DOIUrl":"https://doi.org/10.1093/noajnl/vdaf017","url":null,"abstract":"<p><p>Glioblastoma (GB) is the most aggressive and frequent tumor in the central nervous system and, in humans, represents the worst prognosis for cancer. GB develops a very complex microenvironment, recruiting and interacting with a variety of cells and soluble factors, including immune cells, cytokines, and sex hormones, that contribute to GB survival and progression. Recent evidence has shown a crosstalk between cytokine and sex hormone signaling in GB. This communication could provide GB resistance to treatments and malignancy. Then, how GB orchestrates this communication is a matter of interest. For instance, a critical interaction between tumor necrosis factor-beta (TGF-β) and estrogen receptor signaling has been reported in regulating epithelial-mesenchymal transition, an essential step in GB progression. Furthermore, an inhibition of TGF-β signaling by androgen receptor has been reported to promote GB tumorigenesis in men. Conversely, it has been described that cytokines regulate steroid hormone production in different organs, and this mechanism could be involved in GB development and progression. All these data suggest an intercommunication between the immune and endocrine systems in the tumor microenvironment. Thus, in this review, we focus on explaining the knowledge about this critical intercommunication system and its implication in GB progression.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf017"},"PeriodicalIF":3.7,"publicationDate":"2025-01-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12063100/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144036338","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Neuro-oncology advancesPub Date : 2025-01-23eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf014
Amro H Mohammad, Rawan Sakalla, William Davalan, Miguel Angel Ruiz-Barerra, Sukhdeep Jatana, Roy Khalaf, Hongda Li, Rebecca Aberra, Tariq Al-Saadi, Roberto J Diaz
{"title":"Rise in post-resection neutrophil-to-lymphocyte ratio correlates with decreased survival in glioblastoma patients.","authors":"Amro H Mohammad, Rawan Sakalla, William Davalan, Miguel Angel Ruiz-Barerra, Sukhdeep Jatana, Roy Khalaf, Hongda Li, Rebecca Aberra, Tariq Al-Saadi, Roberto J Diaz","doi":"10.1093/noajnl/vdaf014","DOIUrl":"10.1093/noajnl/vdaf014","url":null,"abstract":"<p><strong>Background: </strong>Neutrophil-to-lymphocyte ratio (NLR) is used in the prognostication of multiple malignancies. However, the NLR value in glioblastoma (GBM) is controversial. This controversy may be due to the unaccounted effect of dexamethasone on NLR. Using retrospective data from 230 isocitrate dehydrogenase-1 (<i>IDH</i>) wild-type GBM patients, we studied the prognostic value of NLR in relation to dexamethasone treatment in GBM.</p><p><strong>Methods: </strong>We retrospectively analyzed 230 patients with GBM. NLR and dexamethasone use were used as dichotomous variables with cutoff values of 9.5 and 8 mg, respectively. Correlations between high NLR, as well as NLR change after surgery, and patient outcome measures, including post-surgical complications and survival, were assessed using Kaplan-Meier curves, logistic, and Cox regression analyses.</p><p><strong>Results: </strong>We demonstrate in this study that high perioperative NLR (≥9.5 NLR) does not associate with survival of GBM patients (274 days, 95% confidence interval [CI] 211-337, vs. 229 days, 95% CI 52-406, <i>P</i> = .9). However, high positive change in NLR (≥6 units) (higher postoperative NLR relative to preoperative NLR) has a significant association with decreased survival in GBM patients (196 days, 95% CI 121-270, vs. 304 days, 95% CI 223-384, <i>P</i> = .01). High preoperative and perioperative average dexamethasone (≥8 mg) treatment did not change the perioperative NLR trend and were not associated with decreased survival.</p><p><strong>Conclusions: </strong>We demonstrate that an increase in NLR after surgery associates with decreased GBM patient survival.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf014"},"PeriodicalIF":3.7,"publicationDate":"2025-01-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11883344/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143573790","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Neuro-oncology advancesPub Date : 2025-01-23eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf016
Su Wang, Rachel Naomi Curry, Malcolm F McDonald, Hyun Yong Koh, Anders W Erickson, Claudia L Kleinman, Michael D Taylor, Ganesh Rao, Benjamin Deneen, Arif O Harmanci, Akdes Serin Harmanci
{"title":"Inferred developmental origins of brain tumors from single-cell RNA-sequencing data.","authors":"Su Wang, Rachel Naomi Curry, Malcolm F McDonald, Hyun Yong Koh, Anders W Erickson, Claudia L Kleinman, Michael D Taylor, Ganesh Rao, Benjamin Deneen, Arif O Harmanci, Akdes Serin Harmanci","doi":"10.1093/noajnl/vdaf016","DOIUrl":"https://doi.org/10.1093/noajnl/vdaf016","url":null,"abstract":"<p><strong>Background: </strong>The reactivation of neurodevelopmental programs in cancer highlights parallel biological processes that occur in both normal development and brain tumors. Achieving a deeper understanding of how dysregulated developmental factors play a role in the progression of brain tumors is therefore crucial for identifying potential targets for therapeutic interventions. Single-cell RNA-sequencing (scRNA-Seq) provides an opportunity to understand how developmental programs are dysregulated and reinitiated in brain tumors at single-cell resolution. The aim of this study is to identify the developmental origins of brain tumors using scRNA-Seq data.</p><p><strong>Methods: </strong>Here, we introduce COORS (Cell Of ORigin like CellS), a computational tool trained on developmental human brain single-cell datasets that annotates \"developmental-like\" cell states in brain tumors. COORS leverages cell type-specific multilayer perceptron models and incorporates a developmental cell type tree that reflects hierarchical relationships and models cell type probabilities.</p><p><strong>Results: </strong>Applying COORS to various brain cancer datasets, including medulloblastoma (MB), glioma, and diffuse midline glioma (DMG), we identified developmental-like cells that represent putative cells of origin in these tumors. Our method provides both cell of origin classification and cell age regression, offering insights into the developmental cell types of tumor subgroups. COORS identified outer radial glia developmental cells within IDH<sup>WT</sup> glioma cells whereas oligodendrocyte precursor cells (OPCs) and neuronal-like cells in IDH<sup>Mut</sup>. Interestingly, IDH<sup>Mut</sup> subgroup cells that map to OPC show bimodal distributions that are both early and late weeks in development. Furthermore, COORS offers a valuable resource by providing novel markers linked to developmental states within MB, glioma, and DMG tumor subgroups.</p><p><strong>Conclusions: </strong>Our work adds to our cumulative understanding of brain tumor heterogeneity and helps pave the way for tailored treatment strategies.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf016"},"PeriodicalIF":3.7,"publicationDate":"2025-01-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12046312/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144036294","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Neuro-oncology advancesPub Date : 2025-01-22eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf015
Metka Novak, Bernarda Majc, Marta Malavolta, Andrej Porčnik, Jernej Mlakar, Matjaž Hren, Anamarija Habič, Mateja Mlinar, Ivana Jovčevska, Neja Šamec, Alja Zottel, Marija Skoblar Vidmar, Tina Vipotnik Vesnaver, Andrej Zupan, Alenka Matjašič, Saša Trkov Bobnar, Dejan Georgiev, Aleksander Sadikov, Roman Bošnjak, Borut Prestor, Radovan Komel, Tamara Lah Turnšek, Barbara Breznik
{"title":"The Slovenian translational platform GlioBank for brain tumor research: Identification of molecular signatures of glioblastoma progression.","authors":"Metka Novak, Bernarda Majc, Marta Malavolta, Andrej Porčnik, Jernej Mlakar, Matjaž Hren, Anamarija Habič, Mateja Mlinar, Ivana Jovčevska, Neja Šamec, Alja Zottel, Marija Skoblar Vidmar, Tina Vipotnik Vesnaver, Andrej Zupan, Alenka Matjašič, Saša Trkov Bobnar, Dejan Georgiev, Aleksander Sadikov, Roman Bošnjak, Borut Prestor, Radovan Komel, Tamara Lah Turnšek, Barbara Breznik","doi":"10.1093/noajnl/vdaf015","DOIUrl":"10.1093/noajnl/vdaf015","url":null,"abstract":"<p><strong>Background: </strong>Glioblastoma (GB) is one of the most lethal solid tumors in humans, with an average patient life expectancy of 15 months and a 5-year survival rate of 5%-10%. GB is still uncurable due to tumor heterogeneity and invasive nature as well as therapy-resistant cancer cells. Centralized biobanks with clinical data and corresponding biological material of GB patients facilitate the development of new treatment approaches and the search for clinically relevant biomarkers, with the goal of improving outcomes for GB patients. The aim of this study was firstly to establish a Slovenian translation platform, GlioBank, and secondly to demonstrate its utility through the identification of molecular signatures associated with GB progression and patient survival.</p><p><strong>Methods: </strong>GlioBank contains tissue samples and corresponding tumor models as well as clinical data from patients diagnosed with glioma, with a focus on GB. Primary GB cells, glioblastoma stem cells (GSCs), and organoids have been established from fresh tumor biopsies. We performed expression analyses of genes associated with GB progression and bioinformatics analyses of available clinical and research data obtained from a subset of 91 GB patients. qPCR was performed to determine the expression of genes associated with therapy resistance and cancer cell invasion, including markers of different GB subtypes, GSCs, epithelial-to-mesenchymal transition, and immunomodulation/chemokine signaling in tumor tissues and corresponding cellular models.</p><p><strong>Results: </strong>GlioBank contains biological material and research, and clinical data collected in the SciNote electronic laboratory notebook. To date, more than 240 glioma tissue samples have been collected and stored in GlioBank, most of which are GB tissues (205) and were further processed to establish primary GB cells (<i>n</i> = 64), GSCs (<i>n</i> = 14), and GB organoids (<i>n</i> = 17). Corresponding blood plasma (<i>n</i> = 103) and peripheral blood mononuclear cells (<i>n</i> = 101) are also stored. GB tumors were classified into 4 different subtypes that differed regarding patient survival; the mixed subtype exhibited the longest patient survival. High <i>DAB2, S100A4</i>, and <i>STAT3</i> expression were associated with poor overall patient survival, and <i>DAB2</i> was found to be an independent prognostic marker for GB survival. We analyzed the molecular signatures between different tumor regions (core vs. rim). <i>STMN4, ERBB3</i>, and <i>ACSBG1</i> were upregulated in the tumor rim, suggesting that these genes are associated with the invasive nature of GB.</p><p><strong>Conclusions: </strong>GlioBank is a centralized biobank that has been built by a multidisciplinary network with the aim of facilitating disease-oriented basic and clinical research. The advantages of GlioBank include the molecular characterization of GB based on targeted gene expression, the availability of diverse cel","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf015"},"PeriodicalIF":3.7,"publicationDate":"2025-01-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11831694/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143443135","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Neuro-oncology advancesPub Date : 2025-01-22eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf004
Michael C LeCompte, Neil Vuppala, Juan M Reyes, Brandi Page, Victoria Croog, Ellen Huang, Kristin J Redmond, Lawrence R Kleinberg
{"title":"Fractionated reirradiation of recurrent high-grade gliomas: Safety with higher reirradiation dose and larger targets.","authors":"Michael C LeCompte, Neil Vuppala, Juan M Reyes, Brandi Page, Victoria Croog, Ellen Huang, Kristin J Redmond, Lawrence R Kleinberg","doi":"10.1093/noajnl/vdaf004","DOIUrl":"10.1093/noajnl/vdaf004","url":null,"abstract":"<p><strong>Background: </strong>The optimal regimen, normal tissue tolerances, and appropriate indications for reirradiation for recurrent high-grade glioma (HGG) are uncertain. The aim of this study was to determine whether higher reirradiation dose was associated with toxicity or survival.</p><p><strong>Methods: </strong>Patients with HGG treated with fractionated reirradiation at a single institution from 2007 to 2022 were retrospectively reviewed. Metrics evaluated included overall survival (OS), prognostic factors for survival, and treatment-related toxicity.</p><p><strong>Results: </strong>Two hundred and thirty patients with recurrent HGG were reviewed. Median follow-up was 8.8 months. Median reirradiation dose was 41.4 Gy with 80.4% receiving concurrent systemic therapy. Median cumulative maximum doses to brainstem and optic structures were 77.9 Gy (range: 4.6-146.0 Gy) and 55.1 Gy (3.3-106.3 Gy), respectively. No injuries to these structures were identified. Radiation necrosis (RN) was identified in 9.4%. There were no significant associations between RN and target size, systemic therapy use, or reirradiation dose. Median OS was 10.2 months from reirradiation start. On multivariate analysis, improved OS was associated with better KPS, longer interval between radiotherapy sessions, reirradiation at first recurrence, and reirradiation dose ≥ 41.4 Gy. Median OS for those with IDH wildtype glioblastoma was 8.7 months. On multivariate analysis of an IDH wildtype disease subanalysis, improved OS was associated with longer interval between radiotherapy sessions and higher reirradiation dose.</p><p><strong>Conclusions: </strong>These data support the safety and efficacy of fractionated reirradiation for recurrent HGG. They suggest higher reirradiation dose may be feasible, including for large treatment volumes and for tumors near the brainstem or optic structures.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf004"},"PeriodicalIF":3.7,"publicationDate":"2025-01-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11842973/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143484891","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Neuro-oncology advancesPub Date : 2025-01-22eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdae231
Alipi V Bonm, Michael S Rutenberg, Kate E Therkelsen, John Herbst, Anurag Sanaf, Marissa A Sherwood, John Y Rhee, Tresa M McGranahan, Patrick J Cimino, L Nicolas Gonzalez Castro, Derek S Tsang, Matthias A Karajannis, Seema Nagpal, Robert J Amdur, Helen Shih, Jason Barber, Lynne P Taylor
{"title":"A multi-institutional retrospective cohort of adult-onset medulloblastoma in the modern era.","authors":"Alipi V Bonm, Michael S Rutenberg, Kate E Therkelsen, John Herbst, Anurag Sanaf, Marissa A Sherwood, John Y Rhee, Tresa M McGranahan, Patrick J Cimino, L Nicolas Gonzalez Castro, Derek S Tsang, Matthias A Karajannis, Seema Nagpal, Robert J Amdur, Helen Shih, Jason Barber, Lynne P Taylor","doi":"10.1093/noajnl/vdae231","DOIUrl":"10.1093/noajnl/vdae231","url":null,"abstract":"<p><strong>Background: </strong>Adult onset medulloblastoma (aMB) is a rare tumor with limited available evidence. We present a large multi-institutional retrospective cohort of aMB patients treated in the modern era, with an emphasis on understanding the role of chemotherapy at initial diagnosis.</p><p><strong>Methods: </strong>We included 267 consecutive patients with aMB treated at 7 different institutions from 2000-present, controlling for chemotherapy regimen and cycles received.</p><p><strong>Results: </strong>Treatment factors were highly intercorrelated with one another and with treating institution. Concurrent chemotherapy was not associated with overall survival (OS). Adjuvant chemotherapy was associated with OS on univariable analyses (HR = 0.55, <i>P</i> = .029) and on multivariable analysis when adjusting for risk status (HR 0.55, <i>P</i> = .026) but not when also adjusting for treating institution. Proton craniospinal irradiation was associated with improved survival on univariable (HR = 0.50, <i>P</i> = .019) and multivariable analysis adjusting for risk status (HR = 0.51, <i>P</i> = .024) but not when treating institution was also considered. On subgroup analysis, adjuvant chemotherapy was associated with improved survival in M0 (HR = 0.55, <i>P</i> = .043) but not M1 disease, in patients with subtotal resection (HR = 0.43, <i>P</i> = .048) but not those with GTR. Similarly, progression-free survival was improved with chemotherapy in patients with M0 (HR = 0.57, <i>P</i> = .032) but not M1 disease, and in patients with subtotal (HR = 0.50, <i>P</i> = .054) but not gross total resection.</p><p><strong>Conclusions: </strong>There was no benefit of concurrent chemotherapy. Adjuvant chemotherapy was associated with improved overall survival and this effect was driven by select subgroups, specifically those with M0 disease and those with residual tumor. We could not confirm that these associations are independent of the treating institution.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdae231"},"PeriodicalIF":3.7,"publicationDate":"2025-01-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11842969/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143484650","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}