Neuro-oncology advancesPub Date : 2025-07-24eCollection Date: 2025-07-01DOI: 10.1093/noajnl/vdaf068
Ruth Lau, Michel Sourour, Alexander Landry, Gelareh Zadeh
{"title":"Honoring the legacy of Dr Fred Gentili, legend of neurosurgery: Pituitary special edition.","authors":"Ruth Lau, Michel Sourour, Alexander Landry, Gelareh Zadeh","doi":"10.1093/noajnl/vdaf068","DOIUrl":"10.1093/noajnl/vdaf068","url":null,"abstract":"","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 Suppl 1","pages":"i1"},"PeriodicalIF":4.1,"publicationDate":"2025-07-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12288124/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144736368","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 : 2025-07-19eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf153
Fair Berg, Gemma Bryan, Allyson Andujar, Dylan E Graetz, Carlos Rodríguez-Galindo, Ibrahim Qaddoumi, Daniel C Moreira
{"title":"Building pediatric neuro-oncology capacity in LMICs through multidisciplinary education and collaboration: a mixed-methods analysis.","authors":"Fair Berg, Gemma Bryan, Allyson Andujar, Dylan E Graetz, Carlos Rodríguez-Galindo, Ibrahim Qaddoumi, Daniel C Moreira","doi":"10.1093/noajnl/vdaf153","DOIUrl":"10.1093/noajnl/vdaf153","url":null,"abstract":"<p><strong>Background: </strong>The St. Jude Global Academy's Neuro-Oncology Training Seminar (NOTS) is a course in pediatric neuro-oncology (PNO), which was created for physicians from low- and middle-income countries. This study reports on the impact of the NOTS.</p><p><strong>Methods: </strong>The NOTS has been imparted in 2018, 2022, and 2023. Data on course participants were prospectively collected, and a survey evaluating course impact was completed by graduates and analyzed quantitatively and qualitatively.</p><p><strong>Results: </strong>Overall, participants from 38 institutions from 29 countries have participated in the NOTS. Furthermore, 205 individuals participated, including 70 pediatric oncologists, 45 neurosurgeons, 22 radiation oncologists, 21 pathologists, 17 radiologists, 7 pediatric neurologists, and 23 other specialties. Survey responses from 76 participants were collected. Of the 47% of respondents who did not have multidisciplinary tumor boards (MDTB) before the NOTS, 55% of those institutions created them influenced by their participation in the NOTS. In addition, 79% of respondents stated that multidisciplinary communication improved after the course. Qualitative analysis identified seven key themes related to the impact of the NOTS course and elucidated the impact of the NOTS on multidisciplinary engagement, institutional practice, and patient care. More specifically, respondents described improvements to interpersonal working, co-ordination of care, clinical management, and decision-making. The NOTS participants have continued to be engaged through the Global Alliance in Pediatric Neuro-Oncology (GAPNO).</p><p><strong>Conclusions: </strong>A multidisciplinary course on the care of pediatric CNS tumor for resource-limited settings has increased PNO expertise and improved multidisciplinary care. It has galvanized a community of practice to further expand capacity building.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf153"},"PeriodicalIF":4.1,"publicationDate":"2025-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12311929/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144762890","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 : 2025-07-19eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf089
Christopher H Dampier, Niharika Shah, Kristyn Galbraith, Azadeh Ebrahimi, Osorio Lopes Abath Neto, Zied Abdullaev, Sanda Alexandrescu, Felipe Andreiuolo, Terri Armstrong, Tiffany Baker, Sahara Cathcart, Hye-Jung Chung, Patrick J Cimino, Kyle S Conway, Jennifer Cotter, Felipe D'Almeida Costa, Karen Dazelle, Nima Etminam, Sima Esther Ferman, Igor Fernandes, Christina K Ferrone, Ahmed Gilani, Mark Gilbert, Jason Gregory, Courtney Ketchum, Han Sung Lee, Ina Lee, Maria Beatriz S Lopes, Qinwen Mao, Michael S Marshall, Matthew McCord, Stewart G Neill, Jeffrey J Nirschl, Byram H Ozer, Werner Paulus, Marta Penas-Prado, Marco Prinz, Peter Pytel, Martha Quezado, Mark Raffeld, Sharika Rajan, Miriam Ratliff, Guido Reifenberger, Lorraina Robinson, Jens Schittenhelm, Daniel Schrimpf, Omkar Singh, Christian Thomas, Diana Thomas, Jaiyeola Thomas-Ogunniyi, Angus Toland, Rust Turakulov, Rachael Vaubel, Nitin Wadhwani, Jing Wu, Caterina Giannini, Matija Snuderl, Sebastian Brandner, Andreas von Deimling, Kenneth Aldape
{"title":"Molecular, histologic, and clinical characterization of methylation class pleomorphic xanthoastrocytoma: An analysis of 469 tumors.","authors":"Christopher H Dampier, Niharika Shah, Kristyn Galbraith, Azadeh Ebrahimi, Osorio Lopes Abath Neto, Zied Abdullaev, Sanda Alexandrescu, Felipe Andreiuolo, Terri Armstrong, Tiffany Baker, Sahara Cathcart, Hye-Jung Chung, Patrick J Cimino, Kyle S Conway, Jennifer Cotter, Felipe D'Almeida Costa, Karen Dazelle, Nima Etminam, Sima Esther Ferman, Igor Fernandes, Christina K Ferrone, Ahmed Gilani, Mark Gilbert, Jason Gregory, Courtney Ketchum, Han Sung Lee, Ina Lee, Maria Beatriz S Lopes, Qinwen Mao, Michael S Marshall, Matthew McCord, Stewart G Neill, Jeffrey J Nirschl, Byram H Ozer, Werner Paulus, Marta Penas-Prado, Marco Prinz, Peter Pytel, Martha Quezado, Mark Raffeld, Sharika Rajan, Miriam Ratliff, Guido Reifenberger, Lorraina Robinson, Jens Schittenhelm, Daniel Schrimpf, Omkar Singh, Christian Thomas, Diana Thomas, Jaiyeola Thomas-Ogunniyi, Angus Toland, Rust Turakulov, Rachael Vaubel, Nitin Wadhwani, Jing Wu, Caterina Giannini, Matija Snuderl, Sebastian Brandner, Andreas von Deimling, Kenneth Aldape","doi":"10.1093/noajnl/vdaf089","DOIUrl":"10.1093/noajnl/vdaf089","url":null,"abstract":"<p><strong>Background: </strong>Methylation class pleomorphic xanthoastrocytoma (mcPXA) comprises tumors with the DNA methylation signature of classical PXA but with a wider histologic spectrum, including overlap with glioblastoma (GBM).</p><p><strong>Methods: </strong>To clarify the histologic and molecular scope of mcPXA and characterize its clinical behavior, a cohort of 469 tumor samples from 458 patients matching to mcPXA by the DKFZ classifier (v12.6 score ≥0.85) was interrogated.</p><p><strong>Results: </strong>Patient median age was 23 years (range 1-73 years) with a female predominance (259 female/199 male). <i>CDKN2A/B</i> homozygous deletion was observed in 406 of 469 (87%) samples. In samples tested for <i>BRAF</i> p.V600E mutations (<i>n</i> = 279), 240 (86%) harbored the mutation. A chr7+/chr10- pattern was observed in 103 of 469 (22%) samples. Among samples tested for <i>TERT</i> promoter mutations (<i>n</i> = 143), 32 (22%) harbored the mutation. Progression-free and overall survival of patients with mcPXA were comparable to patients with methylation class IDH-mutant astrocytoma, low grade, but a GBM-like subset (ie, cases with a pre-methylation working diagnosis of GBM) showed shorter survival. Histologic features of high grade, including palisading necrosis and microvascular proliferation, were prognostic in mcPXA. Compared to patients with <i>BRAF</i> p.V600E-altered GBM, patients with mcPXA were younger and had a lower frequency of <i>TERT</i> promoter mutations.</p><p><strong>Conclusion: </strong>Tumors in mcPXA share molecular characteristics with histologically defined PXA, and high-grade histologic features can help predict their clinical behavior. The use of an epigenetic classification of PXA reveals that this group of tumors is more common than previously appreciated and warrants in-depth study to identify efficacious therapeutic options.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf089"},"PeriodicalIF":4.1,"publicationDate":"2025-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12305539/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144746669","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 : 2025-07-19eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf159
Aaron Michael Taylor, Jody T Lombardi, Areeba Patel, Ariella Tamariz, Jonathan Martin, Markus J Bookland, David S Hersh, Evan Cantor, Xianyuan Song, Felix Sahm, Patrick Kwok-Shing Ng, Joanna J Gell, Ching C Lau
{"title":"A feasibility study of enzymatic methylation sequencing of cell-free DNA from cerebrospinal fluid of pediatric central nervous system tumor patients for molecular classification.","authors":"Aaron Michael Taylor, Jody T Lombardi, Areeba Patel, Ariella Tamariz, Jonathan Martin, Markus J Bookland, David S Hersh, Evan Cantor, Xianyuan Song, Felix Sahm, Patrick Kwok-Shing Ng, Joanna J Gell, Ching C Lau","doi":"10.1093/noajnl/vdaf159","DOIUrl":"10.1093/noajnl/vdaf159","url":null,"abstract":"<p><strong>Background: </strong>Array-based DNA methylation profiling is the gold standard for central nervous system (CNS) tumor molecular classification, but requires over 100 ng input DNA from surgical tissue. Cell-free tumor DNA (cfDNA) in cerebrospinal fluid (CSF) offers an alternative for diagnosis and disease monitoring. This study aimed to test the utilization of enzymatic DNA methylation sequencing (EM-seq) methods to overcome input DNA limitations.</p><p><strong>Methods: </strong>We used the NEBNext EM-seq v2 kit on various amounts of cfDNA, as low as 0.1 ng, extracted from archival CSF samples of 10 patients with CNS tumors. Tumor classification was performed via MNP-Flex using CpG sites overlapping those on the MethylationEPIC array.</p><p><strong>Results: </strong>EM-seq provided sufficient genomic coverage for 10 and 1 ng input DNA samples to generate global DNA methylation profiles. Samples with 0.1 ng input showed lower coverage due to read duplication. Methylation levels for CpG sites with at least 5× coverage were highly correlated across various input DNA amounts, indicating that lower input cfDNA can still be used for tumor classification. The MNP-Flex classifier, trained on tissue DNA methylation data, successfully predicted CNS tumor types for 7 out of 10 CSF samples using EM-seq methylation data with only 1 ng of input cfDNA, consistent with diagnoses based on tissue MethylationEPIC classification and/or histopathology. Additionally, we detected focal and arm-level copy number alterations previously identified via clinical cytogenetics of tumor tissue.</p><p><strong>Conclusions: </strong>This study demonstrated the feasibility of CNS tumor molecular classification based on CSF using the EM-seq approach, and establishes potential sample quality limitations for future studies.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf159"},"PeriodicalIF":4.1,"publicationDate":"2025-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12311925/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144762888","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 : 2025-07-19eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf128
Evan D Bander, Andrew L A Garton, Luca Pasquini, Anne S Reiner, Onur Yildirim, Ahmet T Ilica, Maria Donzelli, Sofia Haque, Mark M Souweidane
{"title":"Patterns of relapse in diffuse intrinsic pontine glioma after convection-enhanced delivery of <sup>124</sup>I-omburtamab.","authors":"Evan D Bander, Andrew L A Garton, Luca Pasquini, Anne S Reiner, Onur Yildirim, Ahmet T Ilica, Maria Donzelli, Sofia Haque, Mark M Souweidane","doi":"10.1093/noajnl/vdaf128","DOIUrl":"10.1093/noajnl/vdaf128","url":null,"abstract":"<p><strong>Background: </strong>Diffuse intrinsic pontine glioma (DIPG) carries a high mortality rate and lacks effective treatment options with a median overall survival (OS) of 8-12 months. Convection-enhanced delivery (CED) has demonstrated safety in phase I trials, but efficacy is indeterminate. Evaluating anatomic patterns of relapse may aid in determining therapeutic efficacy of local CED drug delivery strategies.</p><p><strong>Methods: </strong>Sixty-three children with DIPG were retrospectively reviewed for first radiographic progression. All patients were treated using conventional external beam radiation (EBRT) and 31 were treated with CED of radiolabeled 124-iodine-omburtamab (NCT01502917). Anatomic patterns of initial progression were coded by independent neuroradiologists. OS and cumulative incidence of progression at each anatomic site were assessed in a competing risk analysis with death as a competing variable and were stratified based on CED treatment.</p><p><strong>Results: </strong>Median OS was 14.67 months for the cohort. Patients receiving CED demonstrated higher rates of progression in general, when considering progression at all anatomical sites (HR 1.79, <i>P</i> = .047); no significant difference was found in OS when stratified by CED treatment (<i>P</i> = .22). However, CED treatment was associated with significantly lower cumulative incidence of local pontine and medullary progression (HR: 0.42, <i>P</i> = .03; HR 0.14, <i>P</i> = .01, respectively) relative to non-CED-treated patients.</p><p><strong>Conclusions: </strong>Anatomically defined patterns of relapse provide evidence for locoregional control in children with DIPG treated with radioimmunotherapy administered by CED. Future CED or local surgical therapy trials can benefit from including detailed patterns of relapse as a prospective outcome.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf128"},"PeriodicalIF":4.1,"publicationDate":"2025-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12288026/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144710410","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}
{"title":"Contrast clearance analysis in neuro-oncology: A systematic review and meta-analysis on differentiating posttreatment changes from tumor progression.","authors":"Mohammadreza Tahamtan, Mahshad Afsharzadeh, Masoumeh Sarvari, Shahryar Rahmani, Mahsa Geravandi, Delaram Amiri, Shahriar Kolahi","doi":"10.1093/noajnl/vdaf161","DOIUrl":"10.1093/noajnl/vdaf161","url":null,"abstract":"<p><strong>Background: </strong>Differentiating tumor progression from posttreatment changes, such as pseudoprogression and radiation necrosis, remains a significant challenge in neuro-oncology. Contrast Clearance Analysis (CCA), or Treatment Response Assessment Maps, has developed as a promising tool for this purpose. This systematic review and meta-analysis evaluate the diagnostic accuracy of CCA in distinguishing tumor progression from treatment-induced changes and compare its performance with other advanced imaging modalities.</p><p><strong>Methods: </strong>Following PRISMA-DTA guidelines, a comprehensive search was conducted across PubMed, Scopus, Web of Science, and Embase up to May 2025. Quality assessment was performed using the QUADAS-2 tool. Diagnostic accuracy metrics, including sensitivity, specificity, and area under the curve (AUC), were pooled using a bivariate random-effects meta-analysis model.</p><p><strong>Results: </strong>Nine studies involving 240 patients and 407 brain lesions were included. Contrast Clearance Analysis demonstrated a pooled sensitivity of 91% (95% CI, 0.84-0.95), a specificity of 92% (95% CI, 0.87-0.95), and an AUC of 88%. Moderate heterogeneity was observed in specificity (<i>I</i>² = 37.3%), with no significant heterogeneity in sensitivity (<i>I</i>² = 0%). Publication bias was detected (<i>P</i> <.001), with the trim-and-fill method suggesting 5 potentially missing studies. Quality assessment revealed a considerable risk of bias in the reference test domain.</p><p><strong>Conclusion: </strong>Contrast Clearance Analysis demonstrates high diagnostic accuracy in differentiating tumor progression from posttreatment changes, outperforming conventional MRI and showing comparable or superior performance to other advanced imaging techniques such as MR perfusion, diffusion-weighted imaging, and MR spectroscopy. However, methodological limitations and variability in reference standards highlight the need for standardized protocols in future research.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf161"},"PeriodicalIF":4.1,"publicationDate":"2025-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12311927/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144762891","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}
{"title":"Bevacizumab alternating chemotherapy for improving the survival of patients with recurrent high-grade glioma.","authors":"Ping-Chuan Liu, Chao-Yang Kuo, Yi-Wei Chen, Chun-Fu Lin, Shih-Chieh Lin, Feng-Chi Chang, Ming-The Chen, Jau-Ching Wu, Yi-Yen Lee","doi":"10.1093/noajnl/vdaf157","DOIUrl":"10.1093/noajnl/vdaf157","url":null,"abstract":"<p><strong>Background: </strong>High-grade glioma (HGG) is an aggressive tumor for which there are no effective therapies at recurrence, especially for isocitrate dehydrogenase (IDH)-wild-type glioblastoma. This retrospective study compared survival outcomes between patients receiving bevacizumab alternating chemotherapy (BAC) and those receiving bevacizumab (BEV) alone.</p><p><strong>Methods: </strong>We collected data from 95 adult patients with rHGG who were treated at our institute between January 2018 and August 2023. The patients were divided into 3 groups based on treatment and glioma grade: BAC regimen to treat grade 3 gliomas (<i>n</i> = 23), BAC regimen to treat grade 4 gliomas (<i>n</i> = 29), and treatment with BEV alone (<i>n</i> = 43). The BAC regimen included 2 cycles of etoposide + carboplatin, followed by 1 cycle of cyclophosphamide + vinblastine, with bevacizumab (10 mg/kg) every 4 weeks. One full cycle lasted approximately 3 months. We analyzed overall survival (OS) and postrecurrence survival (PRS).</p><p><strong>Results: </strong>In patients with grade 4 gliomas, the BAC regimen significantly improved survival compared with BEV alone, with a median OS of 29 versus 19 months and a PRS of 16 versus 10 months (both <i>P</i> < .05). In the IDH-wild-type subgroup, the BAC regimen produced a median OS of 27 versus 19 months and a PRS of 16 versus 10 months (<i>P</i> < .05). The 2-year OS and PRS rates were also higher in the BAC groups. Notably, patients with MGMT-methylated grade 4 gliomas treated with the BAC regimen had the longest median OS, 33 months.</p><p><strong>Conclusions: </strong>The BAC regimen appears effective and well tolerated in adult patients with rHGG, particularly in younger patients. Its alternating design may improve the median OS (29 vs. 19 months) and PRS (16 vs. 10 months) of patients with grade 4 gliomas while maintaining safety. As a practical option for those ineligible for clinical trials, BAC warrants further evaluation in prospective randomized studies to confirm its benefits and address the limitations of retrospective analysis.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf157"},"PeriodicalIF":4.1,"publicationDate":"2025-07-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12311935/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144762889","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}
{"title":"Impact of baseline glucocorticoid use on the efficacy of immunotherapy combined with intracranial radiotherapy in NSCLC patients with brain metastases.","authors":"Jin Xiong, Wenhao Shi, Yusheng Huang, Hongmei Jian, Zaicheng Xu, Hongjun Tang, Shunping Huang, Zhenzhou Yang, Yuan Peng","doi":"10.1093/noajnl/vdaf158","DOIUrl":"10.1093/noajnl/vdaf158","url":null,"abstract":"<p><strong>Background: </strong>Glucocorticoids (GCs) play a crucial therapeutic role in managing cerebral edema caused by radiation-induced acute brain injury or brain metastases. However, they negatively impact the efficacy of immunotherapy.</p><p><strong>Methods: </strong>We collected data from 62 patients with non-small cell lung cancer (NSCLC) brain metastases who received immunotherapy combined with intracranial radiotherapy within 28 days of each other. The overall doses of GCs for each patient were expressed as the cumulative total dose of dexamethasone equivalents during the baseline period of immunotherapy. These patients were categorized based on their baseline GCs usage (28 days before and after the first immunotherapy) into three groups: low GCs use (< 30 mg), medium GCs use (30-100 mg), and high GCs use (≥ 100 mg).</p><p><strong>Results: </strong>Among the three groups of included patients in our study, the median intracranial progression-free survival (iPFS) was significantly shorter in the high GCs use group compared to the medium and low GCs use groups (5.23 months vs. 12.70 months vs. 16.43 months, <i>P</i> < .001). No significant difference in median iPFS was observed between the medium and low GCs use groups. Median overall survival (OS) and median progression-free survival (PFS) among the three groups had a similar trend with iPFS. No significant differences in intracranial objective response rate (iORR) and objective response rate (ORR) were found among the three groups. Standard propensity score matching (PSM) confirmed that the high GCs use group (≥ 100 mg) still had significantly shorter median iPFS, PFS, and OS compared to the other groups.</p><p><strong>Conclusions: </strong>Increased baseline GCs use is associated with reduced efficacy of the combination therapy in these patients and baseline high-dose GCs use (≥ 100 mg) notably impairs survival outcomes.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf158"},"PeriodicalIF":4.1,"publicationDate":"2025-07-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12311932/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144762892","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 : 2025-07-11eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf152
Julia Furtner, Luzia Berchtold, Emilie Le Rhun, Antonio Silvani, Roberta Rudà, Giuseppe Lombardi, Juan Manuel Sepúlveda-Sánchez, Petter Brandal, Martin Bendszus, Vassilis Golfinopoulos, Thierry Gorlia, Felix Sahm, Wolfgang Wick, Giuseppe Minniti, Michael Weller, Franz König, Matthias Preusser
{"title":"Prognostic value of 1D, 2D, and volumetric tumor size increases in recurrent WHO grade 2 and 3 meningiomas: Radiological post-hoc analysis of the EORTC-BTG-1320 trial.","authors":"Julia Furtner, Luzia Berchtold, Emilie Le Rhun, Antonio Silvani, Roberta Rudà, Giuseppe Lombardi, Juan Manuel Sepúlveda-Sánchez, Petter Brandal, Martin Bendszus, Vassilis Golfinopoulos, Thierry Gorlia, Felix Sahm, Wolfgang Wick, Giuseppe Minniti, Michael Weller, Franz König, Matthias Preusser","doi":"10.1093/noajnl/vdaf152","DOIUrl":"10.1093/noajnl/vdaf152","url":null,"abstract":"<p><strong>Background: </strong>Although the differential prognostic value of 1D, 2D, and volumetric meningioma size assessment has been reported, RANO meningioma criteria rely on bidimensional measurements.</p><p><strong>Methods: </strong>In this post-hoc analysis of the EORTC-BTG 1320 trial, contrast-enhancing CNS WHO grade 2 and 3 meningiomas were assessed using 1D, 2D, and volumetric measurements. Different cutoff values for lesion size increase were compared 6 months after the start of antineoplastic treatment using Cox proportional hazards models to evaluate their association with overall survival (OS). Optimal cutoff values were identified using two criteria: maximal hazard ratio (HR) for death with statistical significance for median OS and the cutoff that maximized mean specificity and sensitivity for predicting 1-year OS.</p><p><strong>Results: </strong>Among 57 evaluable patients, unidimensional 5 mm and 10 mm tumor size increase yielded the maximal HRs (HR = 3.41, 95% Confidence Interval (CI) 1.56-7.45, <i>P</i> < .01 and HR = 3.22, 95% CI 1.58-6.58, <i>P</i> < .01, respectively) for OS. A 6 mm tumor size increase maximized mean specificity and sensitivity (HR = 2.91, 95% CI 1.43-5.93, <i>P</i> < .01) for predicting 1-year OS. For tumor volume assessments, a 30% increase was associated with the maximal HR (HR = 3.69, 95% CI 1.64-8.31, <i>P</i> < .01) for OS whereas a 40% increase maximized the mean specificity and sensitivity (HR = 3.66, 95% CI 1.75-7.654, <i>P</i> < .01). Bidimensional measurements showed no significant OS association.</p><p><strong>Conclusion: </strong>Unidimensional tumor measurements and tumor volume assessments show a stronger association with overall survival than bidimensional measurements in recurrent non-benign meningiomas. Integration of these methods into response assessment criteria for meningiomas should be considered.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf152"},"PeriodicalIF":4.1,"publicationDate":"2025-07-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12305537/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144746670","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 : 2025-07-11eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf149
Eli E Bar, Julie-Aurore Losman
{"title":"Exploiting genetic and environmental vulnerabilities to target DHODH in CNS tumors.","authors":"Eli E Bar, Julie-Aurore Losman","doi":"10.1093/noajnl/vdaf149","DOIUrl":"10.1093/noajnl/vdaf149","url":null,"abstract":"<p><p>This review explores innovative therapeutic strategies for treating central nervous system (CNS) tumors by targeting their unique metabolic dependencies. This approach marks a significant departure from traditional cytotoxic treatments, focusing instead on the metabolic vulnerabilities created by the tumor's microenvironment and genetic profile. A key area of interest is the de novo pyrimidine synthesis pathway, which is crucial for DNA and RNA synthesis, DNA repair, and protein glycosylation. We highlight the potential of dihydroorotate dehydrogenase (DHODH) inhibitors, which have shown promising anti-tumor activity in preclinical models. The blood-brain barrier, while a challenge for drug delivery, may enhance the efficacy of these inhibitors by maintaining a unique metabolic environment in the brain. Specific brain tumors, such as glioblastoma multiforme, <i>MYC</i>-amplified medulloblastoma, and <i>IDH</i> mutant gliomas, exhibit heightened sensitivity to DHODH inhibition. We suggest that the unique metabolic environment of the brain could make DHODH a more effective therapeutic target for brain tumors compared to other cancer types. Despite the speculative nature of these findings, the compelling preclinical data warrant further investigation into brain-penetrant DHODH inhibitors for CNS malignancies.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf149"},"PeriodicalIF":3.7,"publicationDate":"2025-07-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12284635/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144700786","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}