Neuro-oncology advancesPub Date : 2025-02-21eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdae213
{"title":"Erratum to: Evolutionary evidence precludes <i>ELP1</i> as a high-penetrance pediatric cancer predisposition syndrome gene.","authors":"","doi":"10.1093/noajnl/vdae213","DOIUrl":"https://doi.org/10.1093/noajnl/vdae213","url":null,"abstract":"<p><p>[This corrects the article DOI: 10.1093/noajnl/vdae165.].</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdae213"},"PeriodicalIF":3.7,"publicationDate":"2025-02-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11843024/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143484783","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 : 2025-02-19eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf022
Benjamin M Ellingson, Qunicy Okobi, Robert Chong, Rhea Plawat, Eva Zhao, Andrei Gafita, Ida Sonni, Saewon Chun, Emese Filka, Jingwen Yao, Donatello Telesca, Shanpeng Li, Gang Li, Albert Lai, Phioanh Nghiemphu, Johannes Czernin, David A Nathanson, Timothy F Cloughesy
{"title":"A comparative study of preclinical and clinical molecular imaging response to EGFR inhibition using osimertinib in glioblastoma.","authors":"Benjamin M Ellingson, Qunicy Okobi, Robert Chong, Rhea Plawat, Eva Zhao, Andrei Gafita, Ida Sonni, Saewon Chun, Emese Filka, Jingwen Yao, Donatello Telesca, Shanpeng Li, Gang Li, Albert Lai, Phioanh Nghiemphu, Johannes Czernin, David A Nathanson, Timothy F Cloughesy","doi":"10.1093/noajnl/vdaf022","DOIUrl":"10.1093/noajnl/vdaf022","url":null,"abstract":"<p><strong>Background: </strong>To demonstrate the potential value of <sup>18</sup>F-fluorodeoxyglucose positron emission tomography (<sup>18</sup>F-FDG PET) as a rapid, non-invasive metabolic imaging surrogate for pharmacological modulation of EGFR signaling in EGFR-driven GBM, we synchronously conducted a preclinical imaging study using patient-derived orthotopic xenograft (PDOX) models and validated it in a phase II molecular imaging study in recurrent GBM (rGBM) patients using osimertinib.</p><p><strong>Methods: </strong>A GBM PDOX mouse model study was performed concurrently with an open-label, single-arm, single-center, phase II study of osimertinib (NCT03732352) that enrolled 12 patients with rGBM with EGFR alterations. Patients received osimertinib daily and 3 <sup>18</sup>F-FDG PET scans: two 24 h apart prior to dosing, and one 48 h after dosing.</p><p><strong>Results: </strong>GBM PDOX models suggest osimertinib has limited impact on both <sup>18</sup>F-FDG uptake (+ 9.8%-+25.9%) and survival (+ 15.5%; <i>P</i> = .01), which may be explained by insufficient exposure in the brain (Kp<sub>uu</sub>: 0.30) required to robustly inhibit the EGFR alterations found in GBM. Treatment with osimertinib had subtle, but measurable decreases in the linear rate of change of <sup>18</sup>F-FDG nSUV growth rate averaging -4.5% per day (<i>P</i> = .01) and change in <sup>18</sup>F-FDG uptake was correlated with change in tumor growth rate (R<sup>2</sup> = 0.4719, <i>P = </i>.0195). No metabolic (PERCIST) or radiographic (RANO) responses were seen, and no improvements in PFS or OS were observed.</p><p><strong>Conclusions: </strong>This study demonstrated the feasibility of using FDG PET as a clinically reliable imaging biomarker for assessing EGFR inhibition in GBM, while revealing osimertinib's limited impact on both metabolic activity and tumor growth in GBM, findings that were concordant between preclinical and clinical observations.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf022"},"PeriodicalIF":3.7,"publicationDate":"2025-02-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11883343/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143575069","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 : 2025-02-16eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf040
Xuan Yu, Yihui Gu, Jun Liu, Jingxuan Huang, Qingfeng Li, Zhichao Wang
{"title":"Emerging mechanism and therapeutic potential of neurofibromatosis type 1-related nerve system tumor: Advancing insights into tumor development.","authors":"Xuan Yu, Yihui Gu, Jun Liu, Jingxuan Huang, Qingfeng Li, Zhichao Wang","doi":"10.1093/noajnl/vdaf040","DOIUrl":"10.1093/noajnl/vdaf040","url":null,"abstract":"<p><p>Neurofibromatosis Type 1 (NF1) is a genetic disorder resulting from mutations in the <i>NF1</i> gene, which increases susceptibility to various nervous system tumors, including plexiform neurofibromas, malignant peripheral nerve sheath tumors, and optic pathway gliomas. Recent research has shown that these tumors are intricately connected to the complex, dynamic interactions within neurons, culminating in neuronal signaling that fosters tumor growth. These interactions offer crucial insights into the molecular mechanisms underpinning tumor development, as well as broader implications for therapeutic strategies. This review summarizes the mechanisms through which mutations in the <i>NF1</i> gene within neural tissues trigger tumorigenesis, while examining the role of the neuron-via factors such as visual experience, neurotransmitter, tumor microenvironment, and psychological influences-in both promoting tumor progression and being affected by the tumors themselves. By investigating the dynamic relationship between NF1-associated nervous system tumor cells and neurons, we aim to shed light on novel biological pathways and disease processes, emphasizing the potential of interdisciplinary approaches that combine neurobiology, oncology, and pharmacology to enhance treatment strategies and even inhibit the tumorigenesis.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf040"},"PeriodicalIF":3.7,"publicationDate":"2025-02-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11934560/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143712601","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 : 2025-02-08eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf033
Solmaz Sahebjam, Raju R Raval, Peter A Forsyth, Heiko Enderling, Nam D Tran, John A Arrington, Robert Macaulay, Haley K Perlow, Joshua D Palmer, Jayeeta Ghose, Prajwal Rajappa, Pierre Giglio, Zihai Li, Arnold B Etame, Sepideh Mokhtari, Ruben J Cruz-Chamorro, Menal Bhandari, Ram Thapa, Timothy J Robinson, Dung-Tsa Chen, Hsiang-Hsuan Michael Yu
{"title":"Phase 1 trial of hypofractionated stereotactic re-irradiation in combination with nivolumab, ipilimumab, and bevacizumab for recurrent high-grade gliomas.","authors":"Solmaz Sahebjam, Raju R Raval, Peter A Forsyth, Heiko Enderling, Nam D Tran, John A Arrington, Robert Macaulay, Haley K Perlow, Joshua D Palmer, Jayeeta Ghose, Prajwal Rajappa, Pierre Giglio, Zihai Li, Arnold B Etame, Sepideh Mokhtari, Ruben J Cruz-Chamorro, Menal Bhandari, Ram Thapa, Timothy J Robinson, Dung-Tsa Chen, Hsiang-Hsuan Michael Yu","doi":"10.1093/noajnl/vdaf033","DOIUrl":"10.1093/noajnl/vdaf033","url":null,"abstract":"<p><strong>Background: </strong>Our previous clinical investigation suggested that hypofractionated stereotactic re-irradiation (HFSRT) and PD-1 blockade may act synergistically to enhance the immune response against glioma. This subsequent trial investigated the dual blockade of CTLA4 and PD-1 in combination with HFSRT and bevacizumab.</p><p><strong>Methods: </strong>This phase I study enrolled eligible patients with bevacizumab-naïve recurrent glioblastoma or anaplastic astrocytoma. Participants received nivolumab, ipilimumab, and bevacizumab concurrently with HFSRT (3000 cGy in 5 fractions). Subsequently, nivolumab, ipilimumab, and bevacizumab were administered for a total of 4 cycles followed by nivolumab and bevacizumab until progression. The primary end point of this study was the safety and tolerability of HFSRT in combination with nivolumab, ipilimumab, and bevacizumab in patients with recurrent HGGs. Secondary end points included 6-month survival and 9-month survival.</p><p><strong>Results: </strong>Twenty-six patients were treated. Treatment-related adverse events (TRAEs) of grade 3 or 4 were observed in 12 (48%) evaluable patients with no unexpected TRAEs. Six months and 9 months survival were 92% (95% CI, 82-100%) and 75% (95% CI, 60-95%), respectively. The median progression-free survival and overall survival were 7.1 months (95% CI, 5.2-12.2) and 15.6 months (95% CI, 11.3-27.0), respectively.</p><p><strong>Conclusions: </strong>The combination of HFSRT with ipilimumab, nivolumab, and bevacizumab is safe. Our results underscore the potential synergies between stereotactic re-irradiation and checkpoint immunotherapy in patients with recurrent high-grade gliomas.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf033"},"PeriodicalIF":3.7,"publicationDate":"2025-02-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11934552/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143712602","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Neuro-oncology advancesPub Date : 2025-01-31eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf026
Sophie H A E Derks, Li Shen Ho, Stephan R Koene, Martijn P A Starmans, Esther Oomen-de Hoop, Arjen Joosse, Maja J A de Jonge, Kishan A T Naipal, Joost L M Jongen, Martin J van den Bent, Marion Smits, Astrid A M van der Veldt
{"title":"Size matters: Early progression of melanoma brain metastases after treatment with immune checkpoint inhibitors.","authors":"Sophie H A E Derks, Li Shen Ho, Stephan R Koene, Martijn P A Starmans, Esther Oomen-de Hoop, Arjen Joosse, Maja J A de Jonge, Kishan A T Naipal, Joost L M Jongen, Martin J van den Bent, Marion Smits, Astrid A M van der Veldt","doi":"10.1093/noajnl/vdaf026","DOIUrl":"10.1093/noajnl/vdaf026","url":null,"abstract":"<p><strong>Background: </strong>Immune checkpoint inhibitors (ICIs) are effective treatments for patients with metastatic melanoma, including patients with brain metastasis (BM). However, half of patients with melanoma BM have intracranial progression within 6 months after the start of ICIs. We investigated whether size affects response to ICIs in patients with melanoma BM.</p><p><strong>Methods: </strong>In this single-center cohort study, patients with melanoma BM who were treated with ICIs between 2012 and 2021 were included. Clinical and radiologic features were collected at baseline. Longest axial diameter of all BMs was measured on baseline and follow-up MRI, and segmentation was performed for all BMs on baseline MRI. Lesion-level logistic regression analysis and patient-level survival analysis were performed for early BM progression (ie, within 6 months after start of ICIs) and intracranial progression-free survival (PFS), respectively.</p><p><strong>Results: </strong>A total of 82 patients were included with a total of 464 BMs. At baseline, 37.8% of patients had ≥ 4 BMs and 53.7% of patients had at least one BM with a diameter ≥ 10 mm. In multivariable analysis on the lesion level, baseline BM diameter was associated with early BM progression (odds ratio 1.10, 95%CI 1.05-1.15, <i>P</i> < .001). On the patient level, having at least one BM ≥ 10mm was associated with shorter intracranial PFS (hazard ratio 2.08, 95%CI 1.64-5.56, <i>P</i> < .001).</p><p><strong>Conclusions: </strong>Large BM diameter was associated with a higher risk of early progression after the start of ICIs. Therefore, local therapy should be considered for patients who are treated with ICIs and who have melanoma BMs ≥ 10 mm.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf026"},"PeriodicalIF":3.7,"publicationDate":"2025-01-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11883345/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143573795","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Neuro-oncology advancesPub Date : 2025-01-29eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf023
Michel Friedrich, Jan-Michael Werner, Joachim P Steinbach, Michael Sabel, Ulrich Herrlinger, Marc Piroth, Gabriele Stoffels, Christian P Filss, Philipp Lohmann, Nadim J Shah, Maximilian I Ruge, Felix M Mottaghy, Roland Goldbrunner, Karl-Josef Langen, Gereon R Fink, Martin Kocher, Norbert Galldiks
{"title":"Functional connectivity between tumor region and resting-state networks as imaging biomarker for overall survival in recurrent gliomas diagnosed by <i>O</i>-(2-[<sup>18</sup>F]fluoroethyl)-l-tyrosine PET.","authors":"Michel Friedrich, Jan-Michael Werner, Joachim P Steinbach, Michael Sabel, Ulrich Herrlinger, Marc Piroth, Gabriele Stoffels, Christian P Filss, Philipp Lohmann, Nadim J Shah, Maximilian I Ruge, Felix M Mottaghy, Roland Goldbrunner, Karl-Josef Langen, Gereon R Fink, Martin Kocher, Norbert Galldiks","doi":"10.1093/noajnl/vdaf023","DOIUrl":"10.1093/noajnl/vdaf023","url":null,"abstract":"<p><strong>Background: </strong>Amino acid PET using the tracer <i>O</i>-(2-[<sup>18</sup>F]fluoroethyl)-l-tyrosine (FET) is one of the most reliable imaging methods for detecting glioma recurrence. Here, we hypothesized that functional MR connectivity between the metabolic active recurrent tumor region and resting-state networks of the brain could serve as a prognostic imaging biomarker for overall survival (OS).</p><p><strong>Methods: </strong>The study included 82 patients (26-81 years; median Eastern Cooperative Oncology Group performance score, 0) with recurrent gliomas following therapy (WHO-CNS 2021 grade 4 glioblastoma, <i>n</i> = 57; grade 3 or 4 astrocytoma, <i>n</i> = 12; grade 2 or 3 oligodendroglioma, <i>n</i> = 13) diagnosed by FET PET simultaneously acquired with functional resting-state MR. Functional connectivity (FC) was assessed between tumor regions and 7 canonical resting-state networks.</p><p><strong>Results: </strong>WHO tumor grade and IDH mutation status were strong predictors of OS after recurrence (<i>P</i> < .001). Overall FC between tumor regions and networks was highest in oligodendrogliomas and was inversely related to tumor grade (<i>P</i> = .031). FC between the tumor region and the dorsal attention network was associated with longer OS (HR, 0.88; 95%CI, 0.80-0.97; <i>P</i> = .007), and showed an independent association with OS (HR, 0.90; 95%CI, 0.81-0.99; <i>P</i> = .033) in a model including clinical factors, tumor volume and MGMT. In the glioblastoma subgroup, tumor volume and FC between the tumor and the visual network (HR, 0.90; 95%CI, 0.82-0.99, <i>P</i> = .031) were independent predictors of survival.</p><p><strong>Conclusions: </strong>Recurrent gliomas exhibit significant FC to resting-state networks of the brain. Besides tumor type and grade, high FC between the tumor and distinct networks could serve as independent prognostic factors for improved OS in these patients.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf023"},"PeriodicalIF":3.7,"publicationDate":"2025-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11904474/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143627338","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Neuro-oncology advancesPub Date : 2025-01-29eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf027
Paul van Schie, Ruben G Huisman, Terry Wiersma, Joost L Knegjens, Edwin P M Jansen, Dieta Brandsma, Annette Compter, Philip C de Witt Hamer, René Post, Gerben R Borst
{"title":"Distant brain failure after stereotactic radiosurgery for brain metastases in patients receiving novel systemic treatments.","authors":"Paul van Schie, Ruben G Huisman, Terry Wiersma, Joost L Knegjens, Edwin P M Jansen, Dieta Brandsma, Annette Compter, Philip C de Witt Hamer, René Post, Gerben R Borst","doi":"10.1093/noajnl/vdaf027","DOIUrl":"10.1093/noajnl/vdaf027","url":null,"abstract":"<p><strong>Background: </strong>Novel systemic therapies, such as immunotherapy and targeted therapies, have shown better systemic disease control in the last decennium. However, the effect of these treatments on distant brain failure (DBF) in patients with brain metastases (BM) remains a topic of discussion. Improving time to DBF leads to longer overall survival (OS), as is reflected in the brain metastasis velocity (BMV). This study presents real world data about the combined effects of local and systemic treatments on DBF and survival.</p><p><strong>Methods: </strong>A retrospective consecutive cohort study was conducted. Patients with newly diagnosed BM were included between June 2018 and May 2020. Factors associated with DBF were analyzed in multivariate models. The association between BMV and overall survival was analyzed with linear regression analysis.</p><p><strong>Results: </strong>Three hundred and three patients were included. Two hundred and sixty-two (86%) patients received stereotactic radiotherapy, 41 (14%) awaited in first instance the intracranial effect of newly started or switched systemic treatment. Median time to DBF after radiotherapy was 21 months (95% CI 15-27), median OS was 20 months (IQR 10-36). Receiving immunotherapy or targeted therapy were associated with a lower hazard of DBF, compared with chemotherapy. The presence of > 5 initial BM and progressive or stable extracranial disease were associated with increased DBF. BMV was significantly associated with overall survival.</p><p><strong>Conclusions: </strong>In this retrospective cohort, patients who received immunotherapy or targeted therapy experienced a reduced risk of DBF in comparison to those treated with chemotherapy. A higher BMV was associated with a decreased OS.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf027"},"PeriodicalIF":3.7,"publicationDate":"2025-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11883346/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143575034","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":"Carboplatin and vinblastine monthly in the optic pathway and hypothalamic gliomas: A retrospective analysis in a single institute.","authors":"Ting-Bin Lin, Chao-Yang Kuo, Feng-Chi Chang, Shih-Chieh Lin, Yi-Wei Chen, Muh-Lii Liang, Yi-Yen Lee","doi":"10.1093/noajnl/vdaf020","DOIUrl":"10.1093/noajnl/vdaf020","url":null,"abstract":"<p><strong>Background: </strong>Chemotherapy plays an important role in the treatment of optic pathway hypothalamic gliomas (OPHGs). Commonly used regimens include carboplatin and vincristine and monotherapy with vinblastine weekly. In this retrospective study, we used a monthly regimen of carboplatin and vinblastine to treat progressive/recurrent OPHGs and evaluated their effectiveness, visual preservation, and toxicity.</p><p><strong>Methods: </strong>The study involved patients with OPGH who were treated with carboplatin and vinblastine once per month. The response, disease progression, overall survival, vision changes, and toxicity were recorded according to their medical charts at our institute, and survival was analyzed.</p><p><strong>Results: </strong>A total of 25 patients were included, including 15 males (60%) and 10 females (40%). The response rate was 11/25 (44%), and the stabilization rate (complete response rate + partial response rate + minor response rate + and stable disease rate) was 21/25 (84%). The 3-year progression-free survival (PFS) rate was 54.6%, and the 5-year PFS rate was 46.8%. The 5-year overall survival rate was 100%. There were 6 patients who showed improved visual acuity (28.6%). Stable vision was found in 52.4% of patients. Only 2 patients experienced severe allergic reactions to carboplatin.</p><p><strong>Conclusions: </strong>The results showed that extending the dosing interval of carboplatin and vinblastine to every month can be seen as a similar response compared with previous regimens. The toxicity of this regimen is milder, and patients benefit from a lower frequency of hospital visits. The regimen can be considered as a choice of the first line of chemotherapy for OPHG patients.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf020"},"PeriodicalIF":3.7,"publicationDate":"2025-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11883347/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143575071","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Neuro-oncology advancesPub Date : 2025-01-29eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdaf024
Eduardo Rodriguez Almaraz, Geno A Guerra, Nadeem N Al-Adli, Jacob S Young, Abraham Dada, Daniel Quintana, Jennie W Taylor, Nancy Ann Oberheim Bush, Jennifer L Clarke, Nicholas A Butowski, John de Groot, Melike Pekmezci, Arie Perry, Andrew W Bollen, Aaron W Scheffler, David V Glidden, Joanna J Phillips, Joseph F Costello, Edward F Chang, Shawn Hervey-Jumper, Mitchel S Berger, Stephen S Francis, Susan M Chang, David A Solomon
{"title":"Longitudinal profiling of IDH-mutant astrocytomas reveals acquired RAS-MAPK pathway mutations associated with inferior survival.","authors":"Eduardo Rodriguez Almaraz, Geno A Guerra, Nadeem N Al-Adli, Jacob S Young, Abraham Dada, Daniel Quintana, Jennie W Taylor, Nancy Ann Oberheim Bush, Jennifer L Clarke, Nicholas A Butowski, John de Groot, Melike Pekmezci, Arie Perry, Andrew W Bollen, Aaron W Scheffler, David V Glidden, Joanna J Phillips, Joseph F Costello, Edward F Chang, Shawn Hervey-Jumper, Mitchel S Berger, Stephen S Francis, Susan M Chang, David A Solomon","doi":"10.1093/noajnl/vdaf024","DOIUrl":"10.1093/noajnl/vdaf024","url":null,"abstract":"<p><strong>Background: </strong>Isocitrate dehydrogenase (IDH)-mutant astrocytomas represent the most frequent primary intraparenchymal brain tumor in young adults, which typically arise as low-grade neoplasms that often progress and transform to higher grade despite current therapeutic approaches. However, the genetic alterations underlying high-grade transformation and disease progression of IDH-mutant astrocytomas remain inadequately defined.</p><p><strong>Methods: </strong>Genomic profiling was performed on 205 IDH-mutant astrocytomas from 172 patients from both initial treatment-naive and recurrent post-treatment tumor specimens. Molecular findings were integrated with clinical outcomes and pathologic features to define the associations of novel genetic alterations in the RAS-MAPK signaling pathway.</p><p><strong>Results: </strong>Likely oncogenic alterations within the RAS-MAPK mitogenic signaling pathway were identified in 13% of IDH-mutant astrocytomas, which involved the <i>KRAS, NRAS, BRAF, NF1</i>, <i>SPRED1</i>, and <i>LZTR1</i> genes. These included focal amplifications and known activating mutations in oncogenic components (e.g. <i>KRAS</i>, <i>BRAF</i>), as well as deletions and truncating mutations in negative regulatory components (e.g. <i>NF1</i>, <i>SPRED1</i>). These RAS-MAPK pathway alterations were enriched in recurrent tumors and occurred nearly always in high-grade tumors, often co-occurring with <i>CDKN2A</i> homozygous deletion. Patients whose IDH-mutant astrocytomas harbored these oncogenic RAS-MAPK pathway alterations had inferior survival compared to those with RAS-MAPK wild-type tumors.</p><p><strong>Conclusions: </strong>These findings highlight novel genetic perturbations in the RAS-MAPK pathway as a likely mechanism contributing to the high-grade transformation and treatment resistance of IDH-mutant astrocytomas that may be a potential therapeutic target for affected patients and used for future risk stratification.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdaf024"},"PeriodicalIF":3.7,"publicationDate":"2025-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11883348/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143575061","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Neuro-oncology advancesPub Date : 2025-01-28eCollection Date: 2025-01-01DOI: 10.1093/noajnl/vdae197
Meryem Abbad Andaloussi, Raphael Maser, Frank Hertel, François Lamoline, Andreas Dominik Husch
{"title":"Exploring adult glioma through MRI: A review of publicly available datasets to guide efficient image analysis.","authors":"Meryem Abbad Andaloussi, Raphael Maser, Frank Hertel, François Lamoline, Andreas Dominik Husch","doi":"10.1093/noajnl/vdae197","DOIUrl":"10.1093/noajnl/vdae197","url":null,"abstract":"<p><strong>Background: </strong>Publicly available data are essential for the progress of medical image analysis, in particular for crafting machine learning models. Glioma is the most common group of primary brain tumors, and magnetic resonance imaging (MRI) is a widely used modality in their diagnosis and treatment. However, the availability and quality of public datasets for glioma MRI are not well known.</p><p><strong>Methods: </strong>In this review, we searched for public datasets of glioma MRI using Google Dataset Search, The Cancer Imaging Archive, and Synapse.</p><p><strong>Results: </strong>A total of 28 datasets published between 2005 and May 2024 were found, containing 62 019 images from 5515 patients. We analyzed the characteristics of these datasets, such as the origin, size, format, annotation, and accessibility. Additionally, we examined the distribution of tumor types, grades, and stages among the datasets. The implications of the evolution of the World Health Organization (WHO) classification on tumors of the brain are discussed, in particular the 2021 update that significantly changed the definition of glioblastoma.</p><p><strong>Conclusions: </strong>Potential research questions that could be explored using these datasets were highlighted, such as tumor evolution through malignant transformation, MRI normalization, and tumor segmentation. Interestingly, only 2 datasets among the 28 studied reflect the current WHO classification. This review provides a comprehensive overview of the publicly available datasets for glioma MRI currently at our disposal, providing aid to medical image analysis researchers in their decision-making on efficient dataset choice.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":"7 1","pages":"vdae197"},"PeriodicalIF":3.7,"publicationDate":"2025-01-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11773385/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143061789","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}