Neuro-oncology advancesPub Date : 2025-04-12eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf074
Ryusuke Hatae, Payal B Watchmaker, Akane Yamamichi, Keith Kyewalabye, Kaori Okada, Su Phyu, Yitzhar Goretsky, Jeffrey Haegelin, Psalm Pineo-Cavanaugh, Marco Gallus, Lan Phung, Tiffany Chen, Haoyu Long, Pavlina Chuntova, David H Raulet, Masaki Terabe, Hideho Okada
{"title":"Comparative evaluation of CAR-expressing T-, NK-, NKT-cells, and macrophages in an immunocompetent mouse glioma model.","authors":"Ryusuke Hatae, Payal B Watchmaker, Akane Yamamichi, Keith Kyewalabye, Kaori Okada, Su Phyu, Yitzhar Goretsky, Jeffrey Haegelin, Psalm Pineo-Cavanaugh, Marco Gallus, Lan Phung, Tiffany Chen, Haoyu Long, Pavlina Chuntova, David H Raulet, Masaki Terabe, Hideho Okada","doi":"10.1093/noajnl/vdaf074","DOIUrl":"10.1093/noajnl/vdaf074","url":null,"abstract":"<p><strong>Background: </strong>While chimeric antigen receptor (CAR) T-cells are promising, there is a rapidly growing interest in developing other CAR-expressing immune cells. However, to date, no reported studies evaluated these cells side-by-side in immune-competent glioma models.</p><p><strong>Methods: </strong>We developed a novel C57BL/6-background transgenic mouse strain with all hematopoietic cells carrying the anti-epidermal growth factor receptor (EGFR)vIII-CAR downstream of a Lox-Stop-Lox cassette in the Rosa26 locus. Crossing with mice transgenic for Vav-Cre allowed the expression of anti-EGFRvIII CAR in all hematopoietic cells. In particular, we evaluated CAR-T, CAR-NKT, CAR-NK-cells, and CAR-macrophages in a syngeneic mouse SB28EGFRVIII glioma model.</p><p><strong>Results: </strong>CAR-NK and CAR-NKT-cells demonstrated anti-tumor effects comparable to CAR-T cells in vitro. A single intratumoral administration of CAR-T and CAR-NKT cells in combination mediated superior therapeutic efficacy compared to CAR-T cells or CAR-NKT-cells alone. A single intravenous infusion of CAR-NK cells following lymphodepletion failed to mediate significant anti-glioma effects. Additionally, intratumoral injection of CAR-NK cells did not confer therapeutic benefit. Contrary to previous reports using human macrophages, CAR-macrophages did not demonstrate enhanced antigen-presentation activity against glioma cells compared to non-CAR macrophages. Intratumorally administered CAR-macrophages failed to demonstrate local persistence or anti-tumor effects in vivo.</p><p><strong>Conclusions: </strong>These data provide a valuable basis as to which immune cells can mediate effective anti-glioma response in an immuno-competent glioma environment. Our data also suggest that a combination of CAR-T and CAR-NKT-cells may represent a promising therapeutic strategy.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf074"},"PeriodicalIF":3.7,"publicationDate":"2025-04-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12080550/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144083043","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 : 2025-04-11eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf072
Christianne V Mojica, Thiago Pimentel Muniz, Xin Wang, Stephanie Baker, Kim Edelstein, Cheryl Kanter, Katherine Mileski, Candice Nguyen, Angela Sekely, Derek S Tsang, Cynthia Hawkins, Uri Tabori, Warren P Mason, Julie Bennett
{"title":"Multidisciplinary adolescent and young adult neuro-oncology clinic: Clinical cases, practice challenges, and future perspectives.","authors":"Christianne V Mojica, Thiago Pimentel Muniz, Xin Wang, Stephanie Baker, Kim Edelstein, Cheryl Kanter, Katherine Mileski, Candice Nguyen, Angela Sekely, Derek S Tsang, Cynthia Hawkins, Uri Tabori, Warren P Mason, Julie Bennett","doi":"10.1093/noajnl/vdaf072","DOIUrl":"https://doi.org/10.1093/noajnl/vdaf072","url":null,"abstract":"<p><strong>Background: </strong>The distinct tumor histopathology, molecular features, and psychosocial needs among adolescents and young adults (AYA) with brain tumors pose challenges within traditional healthcare systems. Establishing a multidisciplinary AYA neuro-oncology clinic has been proposed to address these gaps in care. This is the first study to describe the framework and patient profile of a multidisciplinary AYA neuro-oncology clinic in a quaternary cancer center in Canada.</p><p><strong>Methods: </strong>Clinic framework was outlined and patients seen from December 2022 to June 2024 were included. Demographic profiles, tumor characteristics, treatment details, clinical trial enrollment, and allied health referrals were collected. Barriers encountered were summarized.</p><p><strong>Results: </strong>The clinic is composed of specialists in pediatric and adult neuro-oncology with seamless referrals to neurosurgery, radiation oncology, and allied health teams. A total of 100 patients (males 54%, females 46%) were seen with a median age of 24 years. Pediatric-type low-grade glioma (PLGG) was the leading diagnosis. BRAF alterations were the primary molecular drivers. Twenty-nine patients received active neuro-oncology management in the clinic. Overall, 77 patients underwent at least one surgery, 31 patients received radiotherapy, and 43 patients received chemotherapy. Trametinib was the primary targeted treatment prescribed. Three patients were eligible and enrolled in clinical trials. Barriers identified included a lack of peer support groups and a paucity of available clinical trials.</p><p><strong>Conclusions: </strong>This study provides insight into the clinical profile of patients seen in a multidisciplinary AYA neuro-oncology clinic in Canada. Multidisciplinary care is feasible and integral in addressing the multifaceted needs of AYAs with brain tumors.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf072"},"PeriodicalIF":3.7,"publicationDate":"2025-04-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12063081/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144001065","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 : 2025-03-27eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf067
Mihir Gupta, Sathish Prabu Sathyamangalam Samiappan, Hasan Alanya, Kanat Yalcin, Batur Gultekin, Radwa Sharaf, Aladine A Elsamadicy, Diego Samper Figuera, Natalia Samuel, Ketu Mishra-Gorur, Luis Kolb, Declan McGuone, Murat Gunel, E Zeynep Erson-Omay, Jennifer Moliterno, Ehud Mendel
{"title":"Somatic SMARCB1 mutation in spinal meningioma represents branched evolution in a patient with multiple sporadic meningiomas.","authors":"Mihir Gupta, Sathish Prabu Sathyamangalam Samiappan, Hasan Alanya, Kanat Yalcin, Batur Gultekin, Radwa Sharaf, Aladine A Elsamadicy, Diego Samper Figuera, Natalia Samuel, Ketu Mishra-Gorur, Luis Kolb, Declan McGuone, Murat Gunel, E Zeynep Erson-Omay, Jennifer Moliterno, Ehud Mendel","doi":"10.1093/noajnl/vdaf067","DOIUrl":"10.1093/noajnl/vdaf067","url":null,"abstract":"","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf067"},"PeriodicalIF":3.7,"publicationDate":"2025-03-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12081948/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144096544","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 : 2025-03-27eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf064
Michel Fabbro, Muriel Thomaso, Amélie Darlix, Virginie Perotin, Caroline Gallay, Marie Charissoux, Anne-Chantal Granier, Nabila Bouazza, Patrice Champoiral, Louise Coutant, Marta Jarlier, Estelle Guerdoux
{"title":"Integrated early palliative care for patients with newly diagnosed glioblastoma: The GLIOSUPPORT feasibility study.","authors":"Michel Fabbro, Muriel Thomaso, Amélie Darlix, Virginie Perotin, Caroline Gallay, Marie Charissoux, Anne-Chantal Granier, Nabila Bouazza, Patrice Champoiral, Louise Coutant, Marta Jarlier, Estelle Guerdoux","doi":"10.1093/noajnl/vdaf064","DOIUrl":"https://doi.org/10.1093/noajnl/vdaf064","url":null,"abstract":"<p><strong>Background: </strong>Early palliative care improves the quality of life (QoL) and survival in patients with cancer; however, its effects in patients with glioblastoma remain unclear. The GLIOSUPPORT study assessed the feasibility (adherence; primary objective) of an early palliative care program integrated into the standard glioblastoma care pathway. Secondary objectives included the description of the patients' characteristics, QoL, and neuropsychological changes over time, end-of-life decisions, end-of-life treatments, and family carers' perceptions/experiences.</p><p><strong>Methods: </strong>This interventional, prospective, longitudinal, feasibility study was conducted in a French comprehensive cancer center. Thirty-five patients with newly diagnosed glioblastoma were required to reach an adherence rate of 60%. Adherence was defined as going to 3 palliative care visits scheduled every 12 weeks. Baseline characteristics were compared in patients who did and did not adhere to the palliative care program. Minimal clinically important differences and cut-offs were used to quantify QoL changes.</p><p><strong>Results: </strong>The adherence rate was 60% (95% CI [42.1%-76.1%]), indicating that the program was feasible. Visual disturbances, communication/initiation deficits, and anxiety were more frequent in the group that did not adhere to the program. Emotional and social functioning, pain, appetite loss, constipation, and headache increased over time (clinically significant differences), whereas neuropsychological disturbances did not change. Half of the participants identified a family proxy and 8.6% wrote advance directives. One month before death, 28.6% of patients were receiving cancer treatment.</p><p><strong>Conclusions: </strong>Integrating early palliative care in glioblastoma management is feasible. The potential benefits on QoL, mood, and survival must now be evaluated in a larger randomized controlled trial.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf064"},"PeriodicalIF":3.7,"publicationDate":"2025-03-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12063082/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144060680","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 : 2025-03-25eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf060
Ashish Wadekar, Hemlata Jangir, Saumya Sahu, Charli Roy, Sumanta Das, Ashish Suri, Mehar Chand Sharma, Chitra Sarkar, Vaishali Suri
{"title":"Evaluating the efficacy of Hip1R, Vimentin, and H3K27me3 as surrogate markers for 1p/19q co-deletion in oligodendrogliomas.","authors":"Ashish Wadekar, Hemlata Jangir, Saumya Sahu, Charli Roy, Sumanta Das, Ashish Suri, Mehar Chand Sharma, Chitra Sarkar, Vaishali Suri","doi":"10.1093/noajnl/vdaf060","DOIUrl":"https://doi.org/10.1093/noajnl/vdaf060","url":null,"abstract":"<p><strong>Background: </strong>Assessment of the chromosomal 1p/19q status is essential to distinguish between IDH-mutant astrocytoma and oligodendroglioma. Genetic analyses, however, are expensive, time-consuming, and not widely accessible. Immunohistochemical loss of ATRX is currently the only established surrogate marker for a non-1p/19q co-deleted genotype. To find cost-effective approaches and improve risk stratification, we aimed to assess the immunohistochemical expression HIP1R, Vimentin, and H3K27me3 as surrogate markers in predicting 1p/19q status for oligodendrogliomas.</p><p><strong>Methods: </strong>A total of 182 adult-type-diffuse gliomas were analyzed for IDH1 R132H, ATRX, P 53, MIB-1-LI, HIP1R, Vimentin, and H3K27me3 expression using immunohistochemistry. 1p/19q co-deletion was assessed by fluorescence in situ hybridization (FISH) assay. IDH sequencing was performed in IDH1 R132H negative cases. Histomorphological and molecular classification of these gliomas was performed according to World Health Organization (WHO) 2021 CNS5 classification.</p><p><strong>Results: </strong>In this study, 102 IDH-mutant oligodendrogliomas, 44 IDH-mutant astrocytomas, and 36 IDH-wild-type glioblastomas exhibited distinct patterns of the IHC markers. In oligodendrogliomas, HIP1R showed either homogeneous or homogenous with mosaic staining, Vimentin was negative and H3K27me3 was lost in all cases. In Astrocytoma and glioblastomas, HIP1R was predominantly mosaic, Vimentin was widely positive, and H3K27me3 was variable. Combining these markers, especially the positivity of HIP1R, negative Vimentin, and complete loss of H3K27me3, achieved perfect diagnostic accuracy, making them highly reliable for differentiating oligodendroglioma from astrocytoma, and glioblastomas.</p><p><strong>Conclusions: </strong>The study demonstrates that the combinations of the three immunohistochemical markers HIP1R, Vimentin, and H3K27me3 can accurately predict 1p/19q co-deletion status in IDH-mutant gliomas. This method offers a reliable, robust, cost-effective alternative to complex techniques like FISH.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf060"},"PeriodicalIF":3.7,"publicationDate":"2025-03-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12063079/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144052530","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 : 2025-03-22eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf058
Iyad Alnahhas, Mehak Majid Khan, Wenyin Shi
{"title":"What single-cell RNA sequencing taught us about <i>MGMT</i> expression in glioblastoma.","authors":"Iyad Alnahhas, Mehak Majid Khan, Wenyin Shi","doi":"10.1093/noajnl/vdaf058","DOIUrl":"https://doi.org/10.1093/noajnl/vdaf058","url":null,"abstract":"<p><strong>Background: </strong>The promoter methylation status of O-6-methylguanine-DNA methyltransferase (<i>MGMT</i>p) is an important prognostic marker in GBM. Previous studies showed that the expression of <i>MGMT</i> based on immunohistochemistry did not correlate with survival. This is partly because nontumor cells express <i>MGMT</i>. Single-cell sequencing assesses gene expression in tumor cells specifically.</p><p><strong>Methods: </strong>We used publicly available data from 3 recent single-cell/nucleus sequencing GBM studies that included <i>MGMT</i>p methylation status data for patients to evaluate <i>MGMT</i> expression at the single-cell level.</p><p><strong>Results: </strong>In the CPTAC study, a median of 0.82% and 5.7% of tumor cells expressed MGMT in the <i>MGMT</i>p methylated group and <i>MGMT</i>p unmethylated group, respectively (<i>P</i>-value .001). In the Neftel study, a median of 0.59% and 14.01% of tumor cells expressed MGMT in the <i>MGMT</i>p methylated group and <i>MGMT</i>p unmethylated group, respectively (<i>P</i>-value .01). Three unmethylated samples (out of 16) had <i>MGMT</i> expression <2%. It is unclear if this is due to technical inaccuracies as the Neftel paper did not specify the detection method for <i>MGMT</i>p methylation. Alternatively, the percentage of GBM cells expressing <i>MGMT</i> as a continuous variable may be more relevant than the dichotomous <i>MGMT</i>p status. The Wang study confirmed that <i>MGMT</i> expression can increase or decrease between paired primary and recurrent samples. The gene set enrichment analysis shows that <i>MGMT</i> expressing and negative cells are enriched with mesenchymal and proneural genes, respectively.</p><p><strong>Conclusion: </strong>Single-cell data suggest that <i>MGMT</i> expression falls on a continuous spectrum. A smaller percentage of tumor cells express <i>MGMT</i> when <i>MGMT</i>p is methylated.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf058"},"PeriodicalIF":3.7,"publicationDate":"2025-03-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12063106/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144004234","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 : 2025-03-22eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf063
Kurl Jamora, Adeodatus Vito Nicanor, Ayah Erjan, Marc Vincent Barcelona, Dana Keilty, Michael Yan, Aristotelis Kalyvas, Marc Bernstein, Paul Kongkham, Gelareh Zadeh, Eshetu Atenafu, Srinivas Raman, Alejandro Berlin, Charles Catton, Peter Chung, Barbara-Ann Millar, Normand Laperriere, Tatiana Conrad, David Shultz, Enrique Gutierrez-Valencia
{"title":"Survival outcomes and clinical characteristics of brain metastases from prostate cancer: A single-center analysis.","authors":"Kurl Jamora, Adeodatus Vito Nicanor, Ayah Erjan, Marc Vincent Barcelona, Dana Keilty, Michael Yan, Aristotelis Kalyvas, Marc Bernstein, Paul Kongkham, Gelareh Zadeh, Eshetu Atenafu, Srinivas Raman, Alejandro Berlin, Charles Catton, Peter Chung, Barbara-Ann Millar, Normand Laperriere, Tatiana Conrad, David Shultz, Enrique Gutierrez-Valencia","doi":"10.1093/noajnl/vdaf063","DOIUrl":"10.1093/noajnl/vdaf063","url":null,"abstract":"<p><strong>Background: </strong>Brain metastases (BrM) from prostate cancer (PC) are rare. This study sought to evaluate their prevalence, clinical features, treatment modalities, and survival outcomes.</p><p><strong>Methods: </strong>From a database of BrM patients, we analyzed 28 cases of prostate cancer treated at our center between 2008 and 2023.</p><p><strong>Results: </strong>BrM from PC comprised 0.7% of cases. The majority of patients had high-risk features at PC diagnosis: median prostate-specific antigen (PSA) at diagnosis was 65.5 ng/ml (range: 3.9-784.7 ng/ml), 82% were Gleason grade group ≥ 4, and 68% had perineural invasion (PNI). At BrM diagnosis, 79% were castrate-resistant. Most patients had concurrent metastases, including bone (94%), lymph nodes (63%), or lung (6%). Fifty percent presented with a single brain lesion, and the median Graded Prognostic Assessment (GPA) score was 1.5 (range: 0.5-2.5). Patients commonly had radiographic brain edema (57%) and neurological symptoms (54%), whereas only 7% had seizures. Median overall survival (OS) was 9.4 months (95% CI: 4.8-14.8 months) after BrM diagnosis. An upward trend in OS was observed with higher GPA (<i>P</i> = .07). Treatment modalities, including surgery with adjuvant radiation, stereotactic radiosurgery, and whole brain radiotherapy, showed no significant difference in median OS (9.4, 10.1, and 11.0 months respectively, <i>P</i> = .79). OS did not significantly differ between patients with a single versus multiple BrM or patients with castrate-sensitive versus castrate-resistant PC.</p><p><strong>Conclusion: </strong>BrMs from prostate cancer are rare and predominantly occur in patients with advanced, castrate-resistant disease, often accompanied by other metastases. This analysis enhances our understanding of the disease trajectory and informs treatment discussions.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf063"},"PeriodicalIF":3.7,"publicationDate":"2025-03-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12082812/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144096546","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 : 2025-03-22eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf061
Polly Taylor, Gabrielle Cruickshank, Jack Wildman, George Doyle, Ed Whittaker, Sara Walker, Claire McKeeve, Claire Faulkner, Laura Yarram-Smith, Paul White, Kathreena M Kurian
{"title":"Defining the recommended gray zone in O6-methylguanine-DNA methyltransferase promoter methylation pyrosequencing reporting: A robust, translatable method to implement new EANO guidelines.","authors":"Polly Taylor, Gabrielle Cruickshank, Jack Wildman, George Doyle, Ed Whittaker, Sara Walker, Claire McKeeve, Claire Faulkner, Laura Yarram-Smith, Paul White, Kathreena M Kurian","doi":"10.1093/noajnl/vdaf061","DOIUrl":"https://doi.org/10.1093/noajnl/vdaf061","url":null,"abstract":"<p><strong>Background: </strong>The DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT) may cause resistance of tumor cells to alkylating agents and is a predictive biomarker in high-grade gliomas treated with temozolomide. Recent European Association of Neuro-Oncology (EANO) guidelines recommend internal validation of MGMT methylation cutoffs and reporting of gray zone values. This study aimed to develop a method to derive a gray zone from pyrosequencing MGMT methylation data.</p><p><strong>Methods: </strong>We developed a method to find the optimal gray zone using pyrosequencing MGMT methylation values (CpG sites 72-83) from 308 glioblastoma cases with overall survival data. Each integer below the methylated threshold defined a new possible gray zone and categorization which was used as a variable in a multivariate Cox proportional hazards regression model. The optimal gray zone was selected as the option that had a statistically different survival function from the methylated and unmethylated groups, with the largest log-likelihood ratio test statistic. We applied the method to a validation cohort of 115 glioblastoma cases.</p><p><strong>Results: </strong>Our method successfully identified a gray zone in our development cohort. The following categorization gave 3 distinct survival functions: methylated ≥12% (<i>n</i> = 152 cases), gray zone 5%-12% (<i>n</i> = 43), and unmethylated <5% (<i>n</i> = 113). This categorization was better at predicting survival than the existing categorization (methylated ≥12%, unmethylated <12%). Validating our method showed a sufficient sample size and time to follow up is recommended to apply our method.</p><p><strong>Conclusions: </strong>We have developed a translatable method to identify the optimal MGMT gray zone from pyrosequencing data in line with recent EANO guidelines, to enhance clinical decision-making.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf061"},"PeriodicalIF":3.7,"publicationDate":"2025-03-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12121717/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144183550","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 : 2025-03-19eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf059
Myrthe J A Koppers, Matthijs Monnikhof, Jan Meeldijk, Thijs Koorman, Niels Bovenschen
{"title":"Chimeric antigen receptor-macrophages: Emerging next-generation cell therapy for brain cancer.","authors":"Myrthe J A Koppers, Matthijs Monnikhof, Jan Meeldijk, Thijs Koorman, Niels Bovenschen","doi":"10.1093/noajnl/vdaf059","DOIUrl":"https://doi.org/10.1093/noajnl/vdaf059","url":null,"abstract":"<p><p>Adoptive cell-based therapy utilizing chimeric antigen receptor (CAR)-T technology holds promise in the field of neuro-oncology. Significant progress has been made in enhancing both the efficacy and safety of CAR-T-cell therapies. However, challenges such as the multifaceted immunosuppressive impact of the tumor microenvironment and insufficient CAR-T-cell infiltration into brain tumor sites remain a major hurdles. Emerging novel approaches utilizing CAR-macrophages (CAR-MACs) show potent results for brain tumor immunotherapy. CAR-MACs localize to tumor sites more readily, increase immune cell infiltrates, and demonstrate high antitumor efficacy by effectively eliminating tumor cells through mechanisms such as phagocytosis or efferocytosis. This review discusses the current advancements in CAR-MAC cell therapies for brain cancer, followed by an overview of research on manufacturing CAR-MACs for clinical application. We further highlight the potential future applications of CAR-MACs in combinatory therapies in the treatment of brain tumors.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf059"},"PeriodicalIF":3.7,"publicationDate":"2025-03-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12080554/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144083042","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}