Nico Teske, Antonio Dono, Jacob S Young, Stephanie T Juenger, Gilbert C Youssef, Levin Häni, Tommaso Sciortino, Francesco Bruno, Jorg Dietrich, Christine Y Mau, Michael Weller, Juergen Beck, Shawn Hervey-Jumper, Annette M Molinaro, Susan M Chang, Martin van den Bent, Michael A Vogelbaum, Maximilian I Ruge, Daniel P Cahill, Roberta Rudà, Lorenzo Bello, Stefan J Grau, Oliver Schnell, Raymond Y Huang, Patrick Y Wen, Nitin Tandon, Mitchel S Berger, Joerg-Christian Tonn, Yoshua Esquenazi, Philipp Karschnia
Background: The oncological role of resection in elderly patients with glioblastoma remains controversial. We evaluated the value of resection in patients ≥65 years with (I) newly diagnosed and (II) recurrent glioblastoma by comparing the prognostic relevance of extent of resection to patients <65 years.
Methods: The international RANO resect group retrospectively collected patients with newly diagnosed and recurrent IDH-wildtype glioblastoma from ten neuro-oncological centers. Associations of residual tumor with molecular and clinical markers and survival were analyzed.
Results: 1260 patients with newly diagnosed glioblastoma were identified, including 512 patients ≥65 years. Lower postoperative contrast-enhancing tumor volumes were favorably associated with survival on uni- and multivariate analyses; however, the associations with outcome were more pronounced in younger patients. Only in patients <65 years, supramaximal resection was associated with more favorable survival (40 vs. 20 months, p=0.001). In 310 patients with first recurrence (≥65 years: 92), maximal resection of contrast-enhancing tumor was associated with favorable outcomes, particularly in younger patients. Neither older nor younger patients had favorable outcome associations of supramaximal resection in the recurrent setting. All findings were confirmed in propensity-score-matched analyses to minimize confounding effects of inherent differences in demographic and clinical markers (including second-line treatments) between older and younger patients.
Conclusions: While complete contrast-enhancing tumor resection is prognostic for favorable outcomes in older patients, associations of supramaximal resection with improved outcomes were only retained in younger patients with newly diagnosed disease. Those findings support stratified surgical approaches.
{"title":"Associations of supramaximal resection with outcome in glioblastoma across age groups: a report of the RANO resect group.","authors":"Nico Teske, Antonio Dono, Jacob S Young, Stephanie T Juenger, Gilbert C Youssef, Levin Häni, Tommaso Sciortino, Francesco Bruno, Jorg Dietrich, Christine Y Mau, Michael Weller, Juergen Beck, Shawn Hervey-Jumper, Annette M Molinaro, Susan M Chang, Martin van den Bent, Michael A Vogelbaum, Maximilian I Ruge, Daniel P Cahill, Roberta Rudà, Lorenzo Bello, Stefan J Grau, Oliver Schnell, Raymond Y Huang, Patrick Y Wen, Nitin Tandon, Mitchel S Berger, Joerg-Christian Tonn, Yoshua Esquenazi, Philipp Karschnia","doi":"10.1093/neuonc/noaf239","DOIUrl":"https://doi.org/10.1093/neuonc/noaf239","url":null,"abstract":"<p><strong>Background: </strong>The oncological role of resection in elderly patients with glioblastoma remains controversial. We evaluated the value of resection in patients ≥65 years with (I) newly diagnosed and (II) recurrent glioblastoma by comparing the prognostic relevance of extent of resection to patients <65 years.</p><p><strong>Methods: </strong>The international RANO resect group retrospectively collected patients with newly diagnosed and recurrent IDH-wildtype glioblastoma from ten neuro-oncological centers. Associations of residual tumor with molecular and clinical markers and survival were analyzed.</p><p><strong>Results: </strong>1260 patients with newly diagnosed glioblastoma were identified, including 512 patients ≥65 years. Lower postoperative contrast-enhancing tumor volumes were favorably associated with survival on uni- and multivariate analyses; however, the associations with outcome were more pronounced in younger patients. Only in patients <65 years, supramaximal resection was associated with more favorable survival (40 vs. 20 months, p=0.001). In 310 patients with first recurrence (≥65 years: 92), maximal resection of contrast-enhancing tumor was associated with favorable outcomes, particularly in younger patients. Neither older nor younger patients had favorable outcome associations of supramaximal resection in the recurrent setting. All findings were confirmed in propensity-score-matched analyses to minimize confounding effects of inherent differences in demographic and clinical markers (including second-line treatments) between older and younger patients.</p><p><strong>Conclusions: </strong>While complete contrast-enhancing tumor resection is prognostic for favorable outcomes in older patients, associations of supramaximal resection with improved outcomes were only retained in younger patients with newly diagnosed disease. Those findings support stratified surgical approaches.</p>","PeriodicalId":19377,"journal":{"name":"Neuro-oncology","volume":" ","pages":""},"PeriodicalIF":13.4,"publicationDate":"2025-10-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145368493","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Authors Kyle S Smith, Sandeep K Dhanda, Catherine A Billups, Edgar Sioson, Congyu Lu, Airen Zaldivar Peraza, Karishma Gangwani, Yimei Li, Qian Li, Tong Lin, Jeff M Michalski, Roger J Packer, James M Olson, Sarah E S Leary, Maryam Fouladi, Amar Gajjar, Xin Zhou, Arzu Onar-Thomas, Paul A Northcott, Giles W Robinson
Background: The identification of clinical and molecular heterogeneity in medulloblastoma has produced risk-stratified therapy, but establishing the most effective yet least toxic regimens has remained elusive owing to numerous treatment options. To improve risk-stratification, we performed an integrated analysis from three clinical trials.
Methods: Medulloblastoma patients from ACNS0331/NCT00085735, ACNS0332/NCT00392327, and SJMB03/NCT00085202 were included if they had methylation profiling. Molecular groups [WNT, SHH, Group 3 (G3) and Group 4 (G4)], subgroups, and copy number variations were procured from methylation profiles and mutations from next-generation sequencing. Data was assembled into an interactive portal to capture patient characteristics. Cross-trial comparisons, univariable, and multivariable analyses were conducted and used to derive a risk-stratification schema.
Results: 898 patients (WNT = 131, SHH = 151, G3 = 220, G4 = 396) were included. Progression-free-survival (PFS) distributions among analogous cross-trial cohorts were not different, demonstrating no survival advantage of any one therapy over another. The addition of carboplatin to high-dose craniospinal irradiation (HDCSI) containing regimen was selectively superior in PFS in G3/G4 subgroup 3 (p = 0.048) and G3/G4 subgroup 2 (p = 0.035) to HDCSI regimens without carboplatin. Nine actionable risk-stratified groups were identified consisting of 2 WNT groups (low, high-risk), 3 SHH groups (low-, average-, very-high-risk) and 4 G3/G4 groups (low-, average-, high-, and very-high-risk).
Conclusions: Our integrated cross-trial analysis suggests toxicity can be reduced by eliminating disproportionate differences in therapy in favor of a more uniform treatment backbone. Moreover, we propose and model a risk-classification system that identifies the most appropriate cohorts on which to trial significant dose reductions in craniospinal irradiation or select treatment intensifications.
{"title":"An integrated analysis of three medulloblastoma clinical trials refines risk-stratification approaches for reducing toxicity and improving survival.","authors":"Authors Kyle S Smith, Sandeep K Dhanda, Catherine A Billups, Edgar Sioson, Congyu Lu, Airen Zaldivar Peraza, Karishma Gangwani, Yimei Li, Qian Li, Tong Lin, Jeff M Michalski, Roger J Packer, James M Olson, Sarah E S Leary, Maryam Fouladi, Amar Gajjar, Xin Zhou, Arzu Onar-Thomas, Paul A Northcott, Giles W Robinson","doi":"10.1093/neuonc/noaf250","DOIUrl":"https://doi.org/10.1093/neuonc/noaf250","url":null,"abstract":"<p><strong>Background: </strong>The identification of clinical and molecular heterogeneity in medulloblastoma has produced risk-stratified therapy, but establishing the most effective yet least toxic regimens has remained elusive owing to numerous treatment options. To improve risk-stratification, we performed an integrated analysis from three clinical trials.</p><p><strong>Methods: </strong>Medulloblastoma patients from ACNS0331/NCT00085735, ACNS0332/NCT00392327, and SJMB03/NCT00085202 were included if they had methylation profiling. Molecular groups [WNT, SHH, Group 3 (G3) and Group 4 (G4)], subgroups, and copy number variations were procured from methylation profiles and mutations from next-generation sequencing. Data was assembled into an interactive portal to capture patient characteristics. Cross-trial comparisons, univariable, and multivariable analyses were conducted and used to derive a risk-stratification schema.</p><p><strong>Results: </strong>898 patients (WNT = 131, SHH = 151, G3 = 220, G4 = 396) were included. Progression-free-survival (PFS) distributions among analogous cross-trial cohorts were not different, demonstrating no survival advantage of any one therapy over another. The addition of carboplatin to high-dose craniospinal irradiation (HDCSI) containing regimen was selectively superior in PFS in G3/G4 subgroup 3 (p = 0.048) and G3/G4 subgroup 2 (p = 0.035) to HDCSI regimens without carboplatin. Nine actionable risk-stratified groups were identified consisting of 2 WNT groups (low, high-risk), 3 SHH groups (low-, average-, very-high-risk) and 4 G3/G4 groups (low-, average-, high-, and very-high-risk).</p><p><strong>Conclusions: </strong>Our integrated cross-trial analysis suggests toxicity can be reduced by eliminating disproportionate differences in therapy in favor of a more uniform treatment backbone. Moreover, we propose and model a risk-classification system that identifies the most appropriate cohorts on which to trial significant dose reductions in craniospinal irradiation or select treatment intensifications.</p>","PeriodicalId":19377,"journal":{"name":"Neuro-oncology","volume":" ","pages":""},"PeriodicalIF":13.4,"publicationDate":"2025-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145391585","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Craniopharyngioma presents malignant clinical manifestations with severe symptoms and higher incidence of hypothalamic dysfunction. The pathogenesis of craniopharyngioma remains unclear, which impedes the development of effective treatments and preventive measures. Comprehensively understanding the neoplastic programming and tumoral evolution may contribute to the improvement of prognosis for craniopharyngioma.
Methods: Using the single cell RNA-sequencing and high-resolution spatial transcriptomics analysis on adamantinomatous craniopharyngioma (ACP), papillary craniopharyngioma (PCP) and Rathke cleft cyst (RCC) to depict the cellular hierarchies and neoplastic evolution. Multiplex immunohistochemistry and ex vivo pituitary culture from animal model were used to identify the putative therapeutic targets.
Results: Cellular hierarchy analysis uncovered a more inflammatory tumor microenvironment in PCP compared to ACP. Parallel genomic variation and gene expression patterns were revealed between PCP and RCC, suggesting a continuum of the same disease spectrum that evolutes from RCC to PCP. Furthermore, we identified the disease-type-unique structures composed of various cellular states, including the tumoral progenitors surrounding the fibrovascular cores in PCP and senescence associated secretory phenotype (SASP)-related cellular hierarchy in ACP, respectively. In addition, SASP-related fibroblast growth factor (FGF) signaling was identified as a potential therapeutic target for ACP and inhibition of it led to decreased tumor cell proliferation in murine model.
Conclusions: Our study reveals the neoplastic programming and evolution of craniopharyngioma at single cell and spatial levels. Notably, SASP-related FGF signaling is identified as a potential therapeutic target for ACP and inhibition of it reduces tumor cell proliferation in murine model.
{"title":"Interrogation of the cellular hierarchies reveals neoplastic evolution and therapeutic vulnerability in craniopharyngioma.","authors":"Bin Zhang, Jing Zhang, Zhidan Li, Hui Sheng, Haotai Li, Yu Lu, Xixi Liu, Zhongwen Xu, Yunhang Huang, Chenyan Zhu, Yi Wen, Xuelian He, Liguo Zhang","doi":"10.1093/neuonc/noaf249","DOIUrl":"https://doi.org/10.1093/neuonc/noaf249","url":null,"abstract":"<p><strong>Background: </strong>Craniopharyngioma presents malignant clinical manifestations with severe symptoms and higher incidence of hypothalamic dysfunction. The pathogenesis of craniopharyngioma remains unclear, which impedes the development of effective treatments and preventive measures. Comprehensively understanding the neoplastic programming and tumoral evolution may contribute to the improvement of prognosis for craniopharyngioma.</p><p><strong>Methods: </strong>Using the single cell RNA-sequencing and high-resolution spatial transcriptomics analysis on adamantinomatous craniopharyngioma (ACP), papillary craniopharyngioma (PCP) and Rathke cleft cyst (RCC) to depict the cellular hierarchies and neoplastic evolution. Multiplex immunohistochemistry and ex vivo pituitary culture from animal model were used to identify the putative therapeutic targets.</p><p><strong>Results: </strong>Cellular hierarchy analysis uncovered a more inflammatory tumor microenvironment in PCP compared to ACP. Parallel genomic variation and gene expression patterns were revealed between PCP and RCC, suggesting a continuum of the same disease spectrum that evolutes from RCC to PCP. Furthermore, we identified the disease-type-unique structures composed of various cellular states, including the tumoral progenitors surrounding the fibrovascular cores in PCP and senescence associated secretory phenotype (SASP)-related cellular hierarchy in ACP, respectively. In addition, SASP-related fibroblast growth factor (FGF) signaling was identified as a potential therapeutic target for ACP and inhibition of it led to decreased tumor cell proliferation in murine model.</p><p><strong>Conclusions: </strong>Our study reveals the neoplastic programming and evolution of craniopharyngioma at single cell and spatial levels. Notably, SASP-related FGF signaling is identified as a potential therapeutic target for ACP and inhibition of it reduces tumor cell proliferation in murine model.</p>","PeriodicalId":19377,"journal":{"name":"Neuro-oncology","volume":" ","pages":""},"PeriodicalIF":13.4,"publicationDate":"2025-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145391628","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Sonali Arora, Nicholas Nuechterlein, Matt Jensen, Gregory Glatzer, Philipp Sievers, Srinidhi Varadharajan, Andrey Korshunov, Felix Sahm, Stephen C Mack, Michael D Taylor, Taran Gujral, Eric C Holland
Background: Medulloblastoma and ependymoma are common pediatric central nervous system tumors with significant molecular and clinical heterogeneity. While molecular subgrouping has enabled classification into molecular subtypes, the extent of heterogeneity within these subgroups remains poorly defined.
Methods: We collected bulk RNA sequencing data from 888 medulloblastoma and 370 ependymoma tumors to establish a comprehensive reference landscape. After rigorous batch effect correction, normalization, and dimensionality reduction, we generated a unified landscape to explore gene expression, signaling pathways, RNA fusions, and copy number variations.
Results: Our transcriptional analysis revealed distinct clustering patterns, including two primary ependymoma compartments, EPN-E1 and EPN-E2, each with specific RNA fusions and molecular signatures. In medulloblastoma, we observed precise stratification of Group 3/4 tumors by subtype and in SHH tumors by patient age. We also identified subtype-specific pathways and gene fusions, enriched in each group.
Conclusions: This transcriptomic landscape serves as a resource for biomarker discovery, diagnostic refinement, and prediction of tumor biology and outcome. By enabling projection of new patients' bulk RNA-seq data onto the reference map using nearest neighbor analysis, the framework supports accurate subtype classification. The landscape is publicly available via Oncoscape, an interactive platform for global exploration and application.
{"title":"Integrated transcriptomic landscape of medulloblastoma and ependymoma reveals novel tumor subtype-specific biology.","authors":"Sonali Arora, Nicholas Nuechterlein, Matt Jensen, Gregory Glatzer, Philipp Sievers, Srinidhi Varadharajan, Andrey Korshunov, Felix Sahm, Stephen C Mack, Michael D Taylor, Taran Gujral, Eric C Holland","doi":"10.1093/neuonc/noaf251","DOIUrl":"10.1093/neuonc/noaf251","url":null,"abstract":"<p><strong>Background: </strong>Medulloblastoma and ependymoma are common pediatric central nervous system tumors with significant molecular and clinical heterogeneity. While molecular subgrouping has enabled classification into molecular subtypes, the extent of heterogeneity within these subgroups remains poorly defined.</p><p><strong>Methods: </strong>We collected bulk RNA sequencing data from 888 medulloblastoma and 370 ependymoma tumors to establish a comprehensive reference landscape. After rigorous batch effect correction, normalization, and dimensionality reduction, we generated a unified landscape to explore gene expression, signaling pathways, RNA fusions, and copy number variations.</p><p><strong>Results: </strong>Our transcriptional analysis revealed distinct clustering patterns, including two primary ependymoma compartments, EPN-E1 and EPN-E2, each with specific RNA fusions and molecular signatures. In medulloblastoma, we observed precise stratification of Group 3/4 tumors by subtype and in SHH tumors by patient age. We also identified subtype-specific pathways and gene fusions, enriched in each group.</p><p><strong>Conclusions: </strong>This transcriptomic landscape serves as a resource for biomarker discovery, diagnostic refinement, and prediction of tumor biology and outcome. By enabling projection of new patients' bulk RNA-seq data onto the reference map using nearest neighbor analysis, the framework supports accurate subtype classification. The landscape is publicly available via Oncoscape, an interactive platform for global exploration and application.</p>","PeriodicalId":19377,"journal":{"name":"Neuro-oncology","volume":" ","pages":""},"PeriodicalIF":13.4,"publicationDate":"2025-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145391562","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Adrian B Levine, Julie Bennett, Prabhumallikarjun Patil, Ian Burns, Robert Siddaway, Cyril Li, Joseline Haizel-Cobbina, Mansuba Rana, Richard Yuditskiy, Andrew Son, Yoshiko Nakano, Palak Patel, I-Chen Ho, Michelle Ku, Alexander T Lyons, José E Velázquez Vega, Matthew J Schniederjan, Craig Erker, Chantel Cacciotti, Mariarita Santi, Ernest J Nelson, Sylvia Cheng, Christopher Dunham, Bev Wilson, Karina Black, Frank Van Landeghem, Liana Nobre, David D Eisenstat, Ana S Guerreiro Stücklin, Annette Weiser, Valerie Larouche, Panagiota Giannakouros, Adriana Fonseca, Lane Williamson, Igor L Fernandes, Ashley S Plant-Fox, Adam Fleming, Shawde Campbell, Naureen Mushtaq, Syed Ibrahim Bukhari, Khurram Minhas, Richard T Graham, Scott Raskin, Filip Jadrijevic-Cvrlje, Louise Ludlow, Mary V Macneil, Jean M Mulcahy-Levy, Samantha Demarsh, Kohei Fukuoka, Kai Yamasaki, Tomonari Suzuki, Fumiharu Ohka, Atsufumi Kawamura, Yoshiki Arakawa, Takashi Ishihara, Fumiyuki Yamasaki, Jordan R Hansford, Amanda Luck, Maclean P Nasrallah, Helen Toledano, Roaya M Masoud, Alvaro Lassaletta, Luis Blasco-Santana, John-Paul Kilday, Alisa Talianski, Caroline Davies, James Johnston, Andrew T Hale, Peter B Dirks, James T Rutka, Michael C Dewan, Uri Tabori, Cynthia E Hawkins
Background: Pediatric-type low-grade gliomas (PLGG) are the most common central nervous system (CNS) tumor in children. Many are indolent and have excellent outcomes, however some inexplicably spread throughout the CNS leading to increased morbidity and mortality.
Methods: To better understand this rare and difficult-to-treat entity, as well as the features associated with dissemination in CNS tumors, we assembled a large international cohort (n = 269) of patients with disseminated PLGG with detailed clinical and molecular characterization, including DNA sequencing and methylome profiling.
Results: We identified three subgroups of patients based on the temporal and spatial distribution of dissemination. Tumors with diffuse leptomeningeal spread without a primary tumor mass and those occurring in infants had the worst clinical outcomes. The genetics overlapped substantially with that of non-disseminated PLGG, suggesting that non-genetic mechanisms are an important contributor to dissemination. Therapeutically, targeted RAS/MAPK-pathway inhibition was more effective than conventional chemotherapy as first or second-line treatment.
Conclusion: In sum, this cohort increases our clinical and biological understanding of this rare disease, provides insights for improving patient care, and directs future clinical trials and basic science research.
{"title":"Integrated clinical and molecular landscape of disseminated pediatric low-grade glioma.","authors":"Adrian B Levine, Julie Bennett, Prabhumallikarjun Patil, Ian Burns, Robert Siddaway, Cyril Li, Joseline Haizel-Cobbina, Mansuba Rana, Richard Yuditskiy, Andrew Son, Yoshiko Nakano, Palak Patel, I-Chen Ho, Michelle Ku, Alexander T Lyons, José E Velázquez Vega, Matthew J Schniederjan, Craig Erker, Chantel Cacciotti, Mariarita Santi, Ernest J Nelson, Sylvia Cheng, Christopher Dunham, Bev Wilson, Karina Black, Frank Van Landeghem, Liana Nobre, David D Eisenstat, Ana S Guerreiro Stücklin, Annette Weiser, Valerie Larouche, Panagiota Giannakouros, Adriana Fonseca, Lane Williamson, Igor L Fernandes, Ashley S Plant-Fox, Adam Fleming, Shawde Campbell, Naureen Mushtaq, Syed Ibrahim Bukhari, Khurram Minhas, Richard T Graham, Scott Raskin, Filip Jadrijevic-Cvrlje, Louise Ludlow, Mary V Macneil, Jean M Mulcahy-Levy, Samantha Demarsh, Kohei Fukuoka, Kai Yamasaki, Tomonari Suzuki, Fumiharu Ohka, Atsufumi Kawamura, Yoshiki Arakawa, Takashi Ishihara, Fumiyuki Yamasaki, Jordan R Hansford, Amanda Luck, Maclean P Nasrallah, Helen Toledano, Roaya M Masoud, Alvaro Lassaletta, Luis Blasco-Santana, John-Paul Kilday, Alisa Talianski, Caroline Davies, James Johnston, Andrew T Hale, Peter B Dirks, James T Rutka, Michael C Dewan, Uri Tabori, Cynthia E Hawkins","doi":"10.1093/neuonc/noaf245","DOIUrl":"https://doi.org/10.1093/neuonc/noaf245","url":null,"abstract":"<p><strong>Background: </strong>Pediatric-type low-grade gliomas (PLGG) are the most common central nervous system (CNS) tumor in children. Many are indolent and have excellent outcomes, however some inexplicably spread throughout the CNS leading to increased morbidity and mortality.</p><p><strong>Methods: </strong>To better understand this rare and difficult-to-treat entity, as well as the features associated with dissemination in CNS tumors, we assembled a large international cohort (n = 269) of patients with disseminated PLGG with detailed clinical and molecular characterization, including DNA sequencing and methylome profiling.</p><p><strong>Results: </strong>We identified three subgroups of patients based on the temporal and spatial distribution of dissemination. Tumors with diffuse leptomeningeal spread without a primary tumor mass and those occurring in infants had the worst clinical outcomes. The genetics overlapped substantially with that of non-disseminated PLGG, suggesting that non-genetic mechanisms are an important contributor to dissemination. Therapeutically, targeted RAS/MAPK-pathway inhibition was more effective than conventional chemotherapy as first or second-line treatment.</p><p><strong>Conclusion: </strong>In sum, this cohort increases our clinical and biological understanding of this rare disease, provides insights for improving patient care, and directs future clinical trials and basic science research.</p>","PeriodicalId":19377,"journal":{"name":"Neuro-oncology","volume":" ","pages":""},"PeriodicalIF":13.4,"publicationDate":"2025-10-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145337379","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Rosa Luning, Pim J French, Wouter J F Vanbilloen, Lisa Dobber, Levi Van Hijfte, Raoull Hoogendijk, Martin J Van Den Bent, Reno Debets, Marjolein Geurts
Background: T cell-based immunotherapies have had limited success in glioma thus far. Here, we evaluate the literature on abundance, spatial distribution and phenotypical characteristics of T cells in the tumor micro-environment (TME) of IDH-mutant and IDH-wildtype glioma, with the aim to understand how these measures relate to immunotherapy resistance and to aid the development of immunotherapies for glioma.
Methods: Medline, Embase, Web of Science Core Collection, Google Scholar and the Cochrane Central Register of Controlled Trials were systematically searched up to May 6th, 2025. Out of 4,303 articles screened, 85 studies examining T cell immunity in human glioma were selected. We collected information about tumor subtype, grade, methods, T cell abundance, spatial distribution, phenotypes and prognostic significance.
Results: T cells are present in the glioma TME, but at heterogeneous and generally low densities, especially in IDH-mutant glioma. T cell abundance increases with higher WHO grade and upon recurrence. T cells cluster around blood vessels, especially in IDH-mutant glioma. Glioma-infiltrating T cells largely display a late-differentiated phenotype (CD45RA-CCR7-C62L-), expressing markers that signify sustained antigen activation and exhaustion (PD-1, CTLA-4, TIM-3, LAG-3, CD39 and TIGIT). This phenotype coincides with decreased anti-tumor cytotoxicity and is spatially enriched in the myeloid-rich, hypoxic tumor core. Prognostic significance remains controversial.
Conclusions: T cells in glioma are scarce, generally fully differentiated and functionally inert. Understanding and reinvigorating the deficient T cell response will be essential for successful immunotherapies. Future research should incorporate functional and spatial immune profiling to optimize and personalize immunotherapeutic strategies for glioma patients.
背景:到目前为止,基于T细胞的免疫疗法在胶质瘤中的成功有限。在这里,我们评估了idh突变型和idh野生型胶质瘤中T细胞的丰度、空间分布和表型特征的文献,旨在了解这些指标与免疫治疗耐药性的关系,并为胶质瘤免疫治疗的发展提供帮助。方法:系统检索截至2025年5月6日的Medline、Embase、Web of Science Core Collection、谷歌Scholar和Cochrane Central Register of Controlled Trials。在筛选的4303篇文章中,85篇研究人类胶质瘤中T细胞免疫的研究被选中。我们收集了肿瘤亚型、分级、方法、T细胞丰度、空间分布、表型和预后意义等信息。结果:T细胞存在于胶质瘤TME中,但密度不均匀且普遍较低,特别是在idh突变胶质瘤中。T细胞丰度随WHO分级和复发而增加。T细胞聚集在血管周围,特别是在idh突变胶质瘤中。胶质瘤浸润的T细胞主要表现为晚期分化表型(CD45RA-CCR7-C62L-),表达持续抗原激活和衰竭的标志物(PD-1、CTLA-4、TIM-3、LAG-3、CD39和TIGIT)。这种表型与降低的抗肿瘤细胞毒性一致,并且在富含髓细胞的低氧肿瘤核心中空间富集。预后意义仍有争议。结论:胶质瘤中的T细胞是稀缺的,一般是完全分化和功能惰性的。了解并激活缺陷T细胞反应对于成功的免疫治疗至关重要。未来的研究应结合功能和空间免疫分析来优化和个性化神经胶质瘤患者的免疫治疗策略。
{"title":"T cell immunity in glioma and potential implications for immunotherapy: a systematic review.","authors":"Rosa Luning, Pim J French, Wouter J F Vanbilloen, Lisa Dobber, Levi Van Hijfte, Raoull Hoogendijk, Martin J Van Den Bent, Reno Debets, Marjolein Geurts","doi":"10.1093/neuonc/noaf236","DOIUrl":"https://doi.org/10.1093/neuonc/noaf236","url":null,"abstract":"<p><strong>Background: </strong>T cell-based immunotherapies have had limited success in glioma thus far. Here, we evaluate the literature on abundance, spatial distribution and phenotypical characteristics of T cells in the tumor micro-environment (TME) of IDH-mutant and IDH-wildtype glioma, with the aim to understand how these measures relate to immunotherapy resistance and to aid the development of immunotherapies for glioma.</p><p><strong>Methods: </strong>Medline, Embase, Web of Science Core Collection, Google Scholar and the Cochrane Central Register of Controlled Trials were systematically searched up to May 6th, 2025. Out of 4,303 articles screened, 85 studies examining T cell immunity in human glioma were selected. We collected information about tumor subtype, grade, methods, T cell abundance, spatial distribution, phenotypes and prognostic significance.</p><p><strong>Results: </strong>T cells are present in the glioma TME, but at heterogeneous and generally low densities, especially in IDH-mutant glioma. T cell abundance increases with higher WHO grade and upon recurrence. T cells cluster around blood vessels, especially in IDH-mutant glioma. Glioma-infiltrating T cells largely display a late-differentiated phenotype (CD45RA-CCR7-C62L-), expressing markers that signify sustained antigen activation and exhaustion (PD-1, CTLA-4, TIM-3, LAG-3, CD39 and TIGIT). This phenotype coincides with decreased anti-tumor cytotoxicity and is spatially enriched in the myeloid-rich, hypoxic tumor core. Prognostic significance remains controversial.</p><p><strong>Conclusions: </strong>T cells in glioma are scarce, generally fully differentiated and functionally inert. Understanding and reinvigorating the deficient T cell response will be essential for successful immunotherapies. Future research should incorporate functional and spatial immune profiling to optimize and personalize immunotherapeutic strategies for glioma patients.</p>","PeriodicalId":19377,"journal":{"name":"Neuro-oncology","volume":" ","pages":""},"PeriodicalIF":13.4,"publicationDate":"2025-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145308671","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
David Schiff, Xiaobu Ye, Jing Li, Benjamin M Ellingson, Patrick Y Wen, Tobias Walbert, Jian Campian, L Burt Nabors, Byram H Ozer, Arati Desai, Antonio Omuro, Serena Desideri, Neeraja Danda, Stuart Grossman, Ranjit S Bindra
Background: Preclinical studies demonstrate activity of PARP inhibitors in IDH mutant gliomas. We investigated safety, tolerability, pharmacokinetics, and efficacy of the PARP inhibitor pamiparib in conjunction with metronomic low-dose temozolomide in patients with recurrent IDH mutant (IDHmt) gliomas in a multicenter Phase I/II/window of opportunity study.
Methods: Patients received pamiparib in conjunction with daily temozolomide. Following Phase I determination of MTD, we enrolled two patient cohorts (Arm A, multiple prior chemotherapy regimens; Arm B, single prior regimen) in a two-stage design. Exploratory cohorts examined grade 4 IDHmt patients and intratumoral pharmacokinetics of pamiparib. The primary endpoint was objective radiographic response (ORR) by RANO criteria.
Results: 66 subjects were enrolled. We established pamiparib 60 mg twice daily with temozolomide 20 mg daily as the phase II dose. In non-enhancing and enhancing tumor, pamiparib exhibited an unbound tumor/plasma ratio of 0.92 and 0.98 respectively. 0/15 Arm A and 1/24 Arm B patients achieved a centrally confirmed partial response. Median progression-free survival for Arm A was 5.9 months (95% CI, 1.2-14.8 months), and for Arm B 9.7 months (95% CI, 5.7-21.7 months). Grade 3+ anemia and neutropenia affected 24% and 33% of patients respectively. Twenty-two of 66 patients (33.3%) discontinued study treatment for reasons other than tumor progression.
Conclusion: Pamiparib appeared to achieve sufficient pharmacologically active concentrations in both enhancing and non-enhancing tumors. While some patients achieved prolonged progression-free survival, combination with temozolomide did not produce a meaningful ORR in IDHmt recurrent gliomas. Cumulative hematologic toxicity was substantial and impacted long-term tolerability.
{"title":"Phase I/II and Window-of-Opportunity Study of Pamiparib and Metronomic Temozolomide for Recurrent IDH Mutant Gliomas.","authors":"David Schiff, Xiaobu Ye, Jing Li, Benjamin M Ellingson, Patrick Y Wen, Tobias Walbert, Jian Campian, L Burt Nabors, Byram H Ozer, Arati Desai, Antonio Omuro, Serena Desideri, Neeraja Danda, Stuart Grossman, Ranjit S Bindra","doi":"10.1093/neuonc/noaf246","DOIUrl":"https://doi.org/10.1093/neuonc/noaf246","url":null,"abstract":"<p><strong>Background: </strong>Preclinical studies demonstrate activity of PARP inhibitors in IDH mutant gliomas. We investigated safety, tolerability, pharmacokinetics, and efficacy of the PARP inhibitor pamiparib in conjunction with metronomic low-dose temozolomide in patients with recurrent IDH mutant (IDHmt) gliomas in a multicenter Phase I/II/window of opportunity study.</p><p><strong>Methods: </strong>Patients received pamiparib in conjunction with daily temozolomide. Following Phase I determination of MTD, we enrolled two patient cohorts (Arm A, multiple prior chemotherapy regimens; Arm B, single prior regimen) in a two-stage design. Exploratory cohorts examined grade 4 IDHmt patients and intratumoral pharmacokinetics of pamiparib. The primary endpoint was objective radiographic response (ORR) by RANO criteria.</p><p><strong>Results: </strong>66 subjects were enrolled. We established pamiparib 60 mg twice daily with temozolomide 20 mg daily as the phase II dose. In non-enhancing and enhancing tumor, pamiparib exhibited an unbound tumor/plasma ratio of 0.92 and 0.98 respectively. 0/15 Arm A and 1/24 Arm B patients achieved a centrally confirmed partial response. Median progression-free survival for Arm A was 5.9 months (95% CI, 1.2-14.8 months), and for Arm B 9.7 months (95% CI, 5.7-21.7 months). Grade 3+ anemia and neutropenia affected 24% and 33% of patients respectively. Twenty-two of 66 patients (33.3%) discontinued study treatment for reasons other than tumor progression.</p><p><strong>Conclusion: </strong>Pamiparib appeared to achieve sufficient pharmacologically active concentrations in both enhancing and non-enhancing tumors. While some patients achieved prolonged progression-free survival, combination with temozolomide did not produce a meaningful ORR in IDHmt recurrent gliomas. Cumulative hematologic toxicity was substantial and impacted long-term tolerability.</p>","PeriodicalId":19377,"journal":{"name":"Neuro-oncology","volume":" ","pages":""},"PeriodicalIF":13.4,"publicationDate":"2025-10-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145302272","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Mirela-Diana Ilie, Álvaro Flores-Martínez, Marie Chanal, Maxime Lepetit, Benoit Samson, Ayoub Lehiani, Alexandre Vasiljevic, Laura Chinezu, Emmanuel Jouanneau, David Bernard, Olivier Gandrillon, Franck Picard, Gérald Raverot, Philippe Bertolino
Background: The tumor microenvironment (TME) represents a promising avenue to understand gonadotroph tumors and develop therapeutic tools. Here, we aimed to gain insight into the tumorigenesis mechanisms driven by the gonadotoph TME.
Methods: Single-cell and spatial-omics were combined with histological analysis. Mice engrafted with tumor cells were used for functional validation.
Results: using single-cell and spatial transcriptomic data from gonadotroph tumors and normal tissues, we identified mast cells in the microenvironment of gonadotroph tumors and confirmed their physical and functional interaction with endothelial cells. Quantification of mast cells in 40 patients suggested their pro-tumoral role as tumors relapsing after surgery harbored more mast cells. More interestingly, the distribution of mast cells was associated with the presence of a higher number of blood vessels, with an increased microvessel density (MVD), and with blood vessels with thicker walls. Ligand-receptor network analysis highlighted VEGFA as a modulator of mast/endothelial cell communication, a result confirmed by the identification of intratumoral mast cells expressing VEGFA in mouse and human gonadotroph tumors. Finally, using mice engrafted with gonadotroph tumor cells, we demonstrated that the depletion of mast cells reduces tumor volume through increased apoptosis. These observations were associated with increased hemorrhagic areas and a significant reduction of the number of blood vessels and MVD as evidenced in human gonadotroph tumors.
Conclusion: we demonstrate that mast cells represent a new actor of the gonadotroph TME, and highlight their pro-angiogenic and pro-tumorigenic roles as potential targets for the therapeutic treatment of gonadotroph tumors.
{"title":"Mast cells act as pro-angiogenic and pro-tumorigenic players in pituitary gonadotroph tumors.","authors":"Mirela-Diana Ilie, Álvaro Flores-Martínez, Marie Chanal, Maxime Lepetit, Benoit Samson, Ayoub Lehiani, Alexandre Vasiljevic, Laura Chinezu, Emmanuel Jouanneau, David Bernard, Olivier Gandrillon, Franck Picard, Gérald Raverot, Philippe Bertolino","doi":"10.1093/neuonc/noaf241","DOIUrl":"https://doi.org/10.1093/neuonc/noaf241","url":null,"abstract":"<p><strong>Background: </strong>The tumor microenvironment (TME) represents a promising avenue to understand gonadotroph tumors and develop therapeutic tools. Here, we aimed to gain insight into the tumorigenesis mechanisms driven by the gonadotoph TME.</p><p><strong>Methods: </strong> Single-cell and spatial-omics were combined with histological analysis. Mice engrafted with tumor cells were used for functional validation.</p><p><strong>Results: </strong> using single-cell and spatial transcriptomic data from gonadotroph tumors and normal tissues, we identified mast cells in the microenvironment of gonadotroph tumors and confirmed their physical and functional interaction with endothelial cells. Quantification of mast cells in 40 patients suggested their pro-tumoral role as tumors relapsing after surgery harbored more mast cells. More interestingly, the distribution of mast cells was associated with the presence of a higher number of blood vessels, with an increased microvessel density (MVD), and with blood vessels with thicker walls. Ligand-receptor network analysis highlighted VEGFA as a modulator of mast/endothelial cell communication, a result confirmed by the identification of intratumoral mast cells expressing VEGFA in mouse and human gonadotroph tumors. Finally, using mice engrafted with gonadotroph tumor cells, we demonstrated that the depletion of mast cells reduces tumor volume through increased apoptosis. These observations were associated with increased hemorrhagic areas and a significant reduction of the number of blood vessels and MVD as evidenced in human gonadotroph tumors.</p><p><strong>Conclusion: </strong> we demonstrate that mast cells represent a new actor of the gonadotroph TME, and highlight their pro-angiogenic and pro-tumorigenic roles as potential targets for the therapeutic treatment of gonadotroph tumors.</p>","PeriodicalId":19377,"journal":{"name":"Neuro-oncology","volume":" ","pages":""},"PeriodicalIF":13.4,"publicationDate":"2025-10-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145302149","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Uncommon Territory: The Clinical and Molecular Profile of Metastatic Gliomas.","authors":"Floris P Barthel","doi":"10.1093/neuonc/noaf244","DOIUrl":"https://doi.org/10.1093/neuonc/noaf244","url":null,"abstract":"","PeriodicalId":19377,"journal":{"name":"Neuro-oncology","volume":" ","pages":""},"PeriodicalIF":13.4,"publicationDate":"2025-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145302267","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Advancing intraoperative diagnostics, one (or rather three) mutations at a time.","authors":"Jason T Huse","doi":"10.1093/neuonc/noaf232","DOIUrl":"https://doi.org/10.1093/neuonc/noaf232","url":null,"abstract":"","PeriodicalId":19377,"journal":{"name":"Neuro-oncology","volume":" ","pages":""},"PeriodicalIF":13.4,"publicationDate":"2025-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145302116","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}