Neuro-oncology advancesPub Date : 2024-07-31eCollection Date: 2024-01-01DOI: 10.1093/noajnl/vdae133
Kate Hetherington, Claire E Wakefield, Brittany C McGill, Katherine M Tucker, Mark W Donoghoe, Rebecca Daly, Jacqueline D Hunter, Mandy Ballinger, Noemi A Fuentes-Bolanos, David S Ziegler
{"title":"When genetics and pediatric cancer collide: Understanding and optimizing families' experiences.","authors":"Kate Hetherington, Claire E Wakefield, Brittany C McGill, Katherine M Tucker, Mark W Donoghoe, Rebecca Daly, Jacqueline D Hunter, Mandy Ballinger, Noemi A Fuentes-Bolanos, David S Ziegler","doi":"10.1093/noajnl/vdae133","DOIUrl":"10.1093/noajnl/vdae133","url":null,"abstract":"<p><strong>Background: </strong>Advances in our understanding of the genetic basis of childhood cancer, including primary central nervous system cancers, are improving the diagnosis, treatment, and clinical management of pediatric patients. To effectively translate scientific breakthroughs into enhanced clinical care, it is essential we understand and learn from the experiences of patients, families, and health professionals.</p><p><strong>Methods: </strong>This report summarizes findings from 4 Australian psychosocial substudies exploring the perspectives of patients, parents, clinicians, and scientists participating in research related to childhood cancer genetics. Specifically, these studies focus on the psychosocial impact of germline testing in children, surveillance for children with a cancer predisposition syndrome and the perspectives of healthcare professionals who deliver this testing and surveillance.</p><p><strong>Results: </strong>Data presented highlight some of the opportunities and challenges associated with the changing context of genetic predisposition testing for children, adolescents and yound adults with cancer and illustrate how embedding psychosocial data collection in clinical research can answer important questions in the field and inform the design of patient-centric models of care, resources, and workforce training.</p><p><strong>Conclusions: </strong>By embracing these perspectives, we can ensure that advances in genetic research translate into enhanced family experiences, and, ultimately, improved outcomes for children and young people with cancer, and their families.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11364933/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142116629","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 : 2024-07-29eCollection Date: 2024-01-01DOI: 10.1093/noajnl/vdae130
Josef Pichler, Tatjana Traub-Weidinger, Kurt Spiegl, Larisa Imamovic, Arthur J A T Braat, Tom J Snijders, Joost J C Verhoeff, Patrick Flamen, Libuse Tauchmanova, Colin Hayward, Andreas Kluge
{"title":"Results from a phase I study of 4-<i>l</i>-[131I]iodo-phenylalanine ([<sup>131</sup>I]IPA) with external radiation therapy in patients with recurrent glioblastoma (IPAX-1).","authors":"Josef Pichler, Tatjana Traub-Weidinger, Kurt Spiegl, Larisa Imamovic, Arthur J A T Braat, Tom J Snijders, Joost J C Verhoeff, Patrick Flamen, Libuse Tauchmanova, Colin Hayward, Andreas Kluge","doi":"10.1093/noajnl/vdae130","DOIUrl":"https://doi.org/10.1093/noajnl/vdae130","url":null,"abstract":"<p><strong>Background: </strong>Glioblastoma (GBM), the most common malignant brain tumor, is associated with devastating outcomes. IPAX-1 was a multicenter, open-label, single-arm phase I study to evaluate carrier-added 4-<i>L</i>-[<sup>131</sup>I]iodo-phenylalanine ([<sup>131</sup>I]IPA) plus external radiation therapy (XRT) in recurrent GBM.</p><p><strong>Methods: </strong>A total of 10 adults with recurrent GBM who had received first-line debulking surgery plus radio-chemotherapy, were randomized to a single-dose regimen (1f; <sup>131</sup>I-IPA 2 GBq before XRT); a fractionated parallel dose regimen (3f-p; 3 <sup>131</sup>I-IPA 670 MBq fractions, in parallel with second-line XRT), or a fractionated sequential dose regimen (3f-s; 3 <sup>131</sup>I-IPA 670 MBq fractions before and after XRT). Metabolic tumor responses were determined using O-(2-[<sup>18</sup>F]fluoroethyl)-l-tyrosine positron emission tomography, while single-photon emission computed tomography was used to guide [<sup>131</sup>I]IPA tumor dosimetry.</p><p><strong>Results: </strong>All dose regimens were well tolerated. Organ-absorbed radiation doses in red marrow (0.38 Gy) and kidney (1.28 Gy) confirmed no radiation-based toxicity. Stable disease was observed in 4 of the 9 patients at 3 months post-treatment (3-month follow-up [FU], 1 patient did not reach protocol-mandated end of study), yielding a response rate of 44.4%. At the 3-month FU, 6 patients demonstrated metabolic stable disease. Median progression-free survival was 4.3 months (95% confidence interval [CI]: 3.3-4.5), while median overall survival was 13 months (95% CI: 7.1-27).</p><p><strong>Conclusions: </strong>Single or fractionated doses of [<sup>131</sup>I]IPA plus XRT were associated with acceptable tolerability and specific tumor targeting in patients with recurrent GBM, warranting further investigation.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-07-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11358817/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142116628","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 : 2024-07-29eCollection Date: 2024-01-01DOI: 10.1093/noajnl/vdae132
Rachel W Chan, Wilfred W Lam, Hanbo Chen, Leedan Murray, Beibei Zhang, Aimee Theriault, Ruby Endre, Sangkyu Moon, Patrick Liebig, Pejman J Maralani, Chia-Lin Tseng, Sten Myrehaug, Jay Detsky, Mary Jane Lim-Fat, Katrina Roberto, Daniel Djayakarsana, Bharathy Lingamoorthy, Hatef Mehrabian, Benazir Mir Khan, Arjun Sahgal, Hany Soliman, Greg J Stanisz
{"title":"Is pulsed saturation transfer sufficient for differentiating radiation necrosis from tumor progression in brain metastases?","authors":"Rachel W Chan, Wilfred W Lam, Hanbo Chen, Leedan Murray, Beibei Zhang, Aimee Theriault, Ruby Endre, Sangkyu Moon, Patrick Liebig, Pejman J Maralani, Chia-Lin Tseng, Sten Myrehaug, Jay Detsky, Mary Jane Lim-Fat, Katrina Roberto, Daniel Djayakarsana, Bharathy Lingamoorthy, Hatef Mehrabian, Benazir Mir Khan, Arjun Sahgal, Hany Soliman, Greg J Stanisz","doi":"10.1093/noajnl/vdae132","DOIUrl":"10.1093/noajnl/vdae132","url":null,"abstract":"<p><strong>Background: </strong>Stereotactic radiosurgery (SRS) for the treatment of brain metastases delivers a high dose of radiation with excellent local control but comes with the risk of radiation necrosis (RN), which can be difficult to distinguish from tumor progression (TP). Magnetization transfer (MT) and chemical exchange saturation transfer (CEST) are promising techniques for distinguishing RN from TP in brain metastases. Previous studies used a 2D continuous-wave (ie, block radiofrequency [RF] saturation) MT/CEST approach. The purpose of this study is to investigate a 3D pulsed saturation MT/CEST approach with perfusion MRI for distinguishing RN from TP in brain metastases.</p><p><strong>Methods: </strong>The study included 73 patients scanned with MT/CEST MRI previously treated with SRS or fractionated SRS who developed enhancing lesions with uncertain diagnoses of RN or TP. Perfusion MRI was acquired in 49 of 73 patients. Clinical outcomes were determined by at least 6 months of follow-up or via pathologic confirmation (in 20% of the lesions).</p><p><strong>Results: </strong>Univariable logistic regression resulted in significant variables of the quantitative MT parameter 1/(R<sub>A</sub>·T<sub>2A</sub>), with 5.9 ± 2.7 for RN and 6.5 ± 2.9 for TP. The highest AUC of 75% was obtained using a multivariable logistic regression model for MT/CEST parameters, which included the CEST parameters of AREX<sub>Amide,0.625µT</sub> (<i>P</i> = .013), AREX<sub>NOE,0.625µT</sub> (<i>P</i> = .008), 1/(R<sub>A</sub>·T<sub>2A</sub>) (<i>P</i> = .004), and T<sub>1</sub> (<i>P</i> = .004). The perfusion rCBV parameter did not reach significance.</p><p><strong>Conclusions: </strong>Pulsed saturation transfer was sufficient for achieving a multivariable AUC of 75% for differentiating between RN and TP in brain metastases, but had lower AUCs compared to previous studies that used a block RF approach.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-07-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11364936/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142116557","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}
S. Goldlust, Samuel Singer, Lori Cappello, A. K. Almekkawi, Kangmin D Lee, Anthony C Ingenito, Brett E Lewis, Themba Nyirenda, Hooman Azmi, G. Kaptain
{"title":"Phase 1 Study of Concomitant Tumor Treating Fields and Temozolomide Chemoradiation for Newly Diagnosed Glioblastoma","authors":"S. Goldlust, Samuel Singer, Lori Cappello, A. K. Almekkawi, Kangmin D Lee, Anthony C Ingenito, Brett E Lewis, Themba Nyirenda, Hooman Azmi, G. Kaptain","doi":"10.1093/noajnl/vdae129","DOIUrl":"https://doi.org/10.1093/noajnl/vdae129","url":null,"abstract":"\u0000 \u0000 \u0000 Glioblastoma (GBM) is the most common and aggressive primary brain tumor and has limited effective therapies. Tumor treating fields (TTF; Optune Gio®) is an FDA- approved device with data supporting a significant survival benefit and minimal toxicity when added to maintenance chemotherapy. Uptake in clinic practice is not universal and might improve if a shorter duration of treatment is feasible. This phase 1 trial was designed to determine the safety and preliminary efficacy of TTF concomitant to chemoradiation.\u0000 \u0000 \u0000 \u0000 Patients with newly diagnosed, histologically confirmed GBM were eligible. Following surgery, patients were treated with TTF concomitant to standard chemoradiation. The device continued through two monthly cycles of maintenance temozolomide with imaging and clinical assessments at regular intervals to assess toxicity and response. The primary endpoint was safety and tolerability of combined modality treatment based upon the incidence and severity of adverse events. Secondary endpoints were overall survival (OS) and progression free survival (PFS).\u0000 \u0000 \u0000 \u0000 Thirteen patients were enrolled. Dermatologic adverse events were frequent but limited to grade 1/2. There was only one serious adverse event possibly related to TTF and no patients were unable to complete the prescribed course of multimodality treatment due to TTF -associated toxicity. Twelve patients were evaluable for median and six-month progression free survival which were 8.5 months (mo) and 66.7% respectively. Median and 12 mo overall survival were 16.0 mo and 83.3% respectively.\u0000 \u0000 \u0000 \u0000 TTF can be safely delivered in conjunction with chemoradiation. The potential for a finite TTF course merits further evaluation.\u0000","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-07-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141796535","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Fertility preserving techniques in neuro-oncology patients: A systematic review.","authors":"Maia Osborne-Grinter, Jasleen Kaur Sanghera, Offorbuike Chiamaka Bianca, Chandrasekaran Kaliaperumal","doi":"10.1093/noajnl/vdae124","DOIUrl":"10.1093/noajnl/vdae124","url":null,"abstract":"<p><strong>Background: </strong>Advancements in cancer treatments have enhanced survival rates and quality of life for patients with central nervous system (CNS) tumors. There is growing recognition of the significance of fertility preservation methods. Currently, techniques, including oocyte cryopreservation and sperm cryopreservation are established. Nevertheless, oncologists may exhibit reluctance when referring patients to reproductive specialists. This review aimed to assess the best evidence for fertility preservation techniques used in patients with CNS cancers and evaluate outcomes relating to their success and complications.</p><p><strong>Methods: </strong>Two reviewers performed a search of Pubmed, Embase, Medline, Cochrane, and Google Scholar. Papers were included if they reported at least 1 fertility preservation technique in a neuro-oncology patient. Non-English studies, editorials, animal studies, and guidelines were excluded. Meta-analysis was performed using the random effects model.</p><p><strong>Results: </strong>Sixteen studies containing data from 237 participants (78.8% female) were included in the systematic review and meta-analysis, of whom 110 (46.4%) underwent fertility preservation techniques. All patients (100%) successfully underwent fertility preservation with 1 participant (2.9%) returning to rewarm their oocytes, embryos or sperm. On average, 17.8 oocytes were retrieved with 78%, ultimately being cryopreserved. Five (6.0%) patients successfully conceived 9 healthy-term children after utilizing their cryopreserved sperm, embryos, or oocytes. Moreover, 6 patients successfully conceived naturally or using intrauterine insemination, resulting in 7 healthy-term children.</p><p><strong>Conclusions: </strong>Fertility preservation techniques could offer a safe and effective way for neuro-oncology patients to deliver healthy-term babies following treatment. However, further studies concerning risks, long-term pregnancy outcomes, and cost-effectiveness are needed.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11364935/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142116556","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 : 2024-07-25eCollection Date: 2024-01-01DOI: 10.1093/noajnl/vdae126
Erin E Crotty, Vera A Paulson, Rebecca Ronsley, Nicholas A Vitanza, Amy Lee, Jason Hauptman, Hannah E Goldstein, Christina M Lockwood, Sarah E S Leary, Bonnie L Cole
{"title":"Cerebrospinal fluid liquid biopsy by low-pass whole genome sequencing for clinical disease monitoring in pediatric embryonal tumors.","authors":"Erin E Crotty, Vera A Paulson, Rebecca Ronsley, Nicholas A Vitanza, Amy Lee, Jason Hauptman, Hannah E Goldstein, Christina M Lockwood, Sarah E S Leary, Bonnie L Cole","doi":"10.1093/noajnl/vdae126","DOIUrl":"https://doi.org/10.1093/noajnl/vdae126","url":null,"abstract":"<p><strong>Background: </strong>Liquid biopsy assays that detect cell-free DNA (cfDNA) in cerebrospinal fluid (CSF) are a promising tool for disease monitoring in pediatric patients with primary central nervous system (CNS) tumors. As a compliment to tissue-derived molecular analyses, CSF liquid biopsy has the potential to transform risk stratification, prognostication, and precision medicine approaches.</p><p><strong>Methods: </strong>In this pilot study, we evaluated a clinical pipeline to determine feasibility and sensitivity of low-pass whole genome sequencing (LP-WGS) of CSF-derived cfDNA from patients with CNS embryonal tumors. Thirty-two longitudinal CSF samples collected from 17 patients with molecularly characterized medulloblastoma (12), embryonal tumor with multilayered rosettes (2), CNS embryonal tumor, not elsewhere classified (NEC) (2), and atypical teratoid/rhabdoid tumor (1) were analyzed.</p><p><strong>Results: </strong>Adequate CSF-derived cfDNA for LP-WGS analysis was obtained in 94% of samples (30/32). Copy number variants compatible with neoplasia were detected in 90% (27/30) and included key alterations, such as isodicentric ch17, monosomy 6, and <i>MYCN</i> amplification, among others. Compared to tissue specimens, LP-WGS detected additional aberrations in CSF not previously identified in corresponding primary tumor specimens, suggesting a more comprehensive profile of tumor heterogeneity or evolution of cfDNA profiles over time. Among the 12 CSF samples obtained at initial staging, only 2 (17%) were cytologically positive, compared to 11 (92%) that were copy number positive by LP-WGS.</p><p><strong>Conclusions: </strong>LP-WGS of CSF-derived cfDNA is feasible using a clinical platform, with greater sensitivity for tumor detection compared to conventional CSF cytologic analysis at initial staging. Large prospective studies are needed to further evaluate LP-WGS as a predictive biomarker.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11407906/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142305115","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}
A. S. Youshani, C. Heal, J. X. Lee, M. Younis, H. Maye, M. Bailey, D. Coope, P. D’urso, K. Karabatsou
{"title":"Glioma-related epilepsy following low-grade glioma surgery","authors":"A. S. Youshani, C. Heal, J. X. Lee, M. Younis, H. Maye, M. Bailey, D. Coope, P. D’urso, K. Karabatsou","doi":"10.1093/noajnl/vdae127","DOIUrl":"https://doi.org/10.1093/noajnl/vdae127","url":null,"abstract":"\u0000 \u0000 \u0000 Epileptic seizures commonly burden low-grade glioma (LGG) patients and negatively impact quality of life, neurocognition and general patient health. Anti-seizure medications (ASM) are used to manage seizures, but can result in undesired side effects. Our aim was to report our experience in epilepsy in one of the largest case series of LGG patients (re-classified in accordance with the WHO 2021 classification). Furthermore, we evaluate our post-operative seizure frequency difference between LGG patients that use pre-operative ASMs and ones with no ASMs.\u0000 \u0000 \u0000 \u0000 Data was retrospectively collected from Salford Royal Hospital electronic records and Neuro-oncology database from 2006 until 2022. Descriptive statistics were performed for demographic analysis, while multivariable analysis was used to determine post-operative seizure-free outcomes.\u0000 \u0000 \u0000 \u0000 In total, 257 operations were performed on 206 patients. Post-operatively, 114 patients suffered with seizures and approximately 45.2% of patients developed seizures at 3-12 months post-surgery, with the odds higher in patients on pre-operative ASMs. There was no evidence to suggest a higher post-operative seizure rate in patients undergoing awake craniotomy versus general anaesthetic. The extent of resection (EOR) was inversely related to seizure failure, with gross-total resection showing a statistically significant reduction in seizures in comparison to all other surgical resections.\u0000 \u0000 \u0000 \u0000 In our experience, there is no evidence to suggest a reduced post-operative seizure outcome when prescribing pre-operative ASMs. EOR is an independent prognosticator for post-operative seizure failure with all other variables demonstrating non-significance. Overall, a larger study can investigate the role of ASMs in LGG in greater detail.\u0000","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-07-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141819130","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Neuro-oncology advancesPub Date : 2024-07-18eCollection Date: 2024-01-01DOI: 10.1093/noajnl/vdae065
Jian Ping Jen, Xuanxuan Li, Markand Patel, Huzaifah Haq, Ute Pohl, Santhosh Nagaraju, Victoria Wykes, Paul Sanghera, Colin Watts, Vijay Sawlani
{"title":"Beyond T2-FLAIR mismatch sign in isocitrate dehydrogenase mutant 1p19q non-codeleted astrocytoma: Analysis of tumor core and evolution with multiparametric magnetic resonance imaging.","authors":"Jian Ping Jen, Xuanxuan Li, Markand Patel, Huzaifah Haq, Ute Pohl, Santhosh Nagaraju, Victoria Wykes, Paul Sanghera, Colin Watts, Vijay Sawlani","doi":"10.1093/noajnl/vdae065","DOIUrl":"10.1093/noajnl/vdae065","url":null,"abstract":"<p><strong>Background: </strong>The T2-FLAIR mismatch sign is an imaging correlate for isocitrate dehydrogenase (IDH)-mutant 1p19q non-codeleted astrocytomas. However, it is only seen in a part of the cases at certain stages. Many of the tumors likely lose T2 homogeneity as they grow in size, and become heterogenous. The aim of this study was to investigate the timecourse of T2-FLAIR mismatch sign, and assess intratumoral heterogeneity using multiparametric magnetic resonance imaging techniques.</p><p><strong>Methods: </strong>A total of 128 IDH-mutant gliomas were retrospectively analyzed. Observers blinded to molecular status used strict criteria to select T2-FLAIR mismatch astrocytomas. Pre-biopsy and follow-up standard structural sequences of T2, FLAIR and apparent diffusion coefficient, MR spectroscopy (both single- and multi-voxel techniques), and DSC perfusion were observed.</p><p><strong>Results: </strong>Nine T2-FLAIR mismatch astrocytomas were identified. 7 had MR spectroscopy and perfusion data. The smallest astrocytomas began as rounded T2 homogeneous lesions without FLAIR suppression, and developed T2-FLAIR mismatch during follow-up with falls in NAA and raised Cho/Cr ratio. Larger tumors at baseline with T2-FLAIR mismatch signs developed intratumoral heterogeneity, and showed elevated Cho/Cr ratio and raised relative cerebral blood volume (rCBV). The highest levels of intratumoral Cho/Cr and rCBV changes were located within the tumor core, and this area signifies the progression of the tumors toward high grade.</p><p><strong>Conclusions: </strong>T2-FLAIR mismatch sign is seen at a specific stage in the development of astrocytoma. By assessing the subsequent heterogeneity, MR spectroscopy and perfusion imaging are able to predict the progression of the tumor towards high grade, thereby can assist targeting for biopsy and selective debulking.</p>","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-07-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11275453/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141790684","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}
Chloe Gui, Luxshikka Canthiya, Gelareh Zadeh, S. Suppiah
{"title":"Current state of spinal nerve sheath tumor management and future advances","authors":"Chloe Gui, Luxshikka Canthiya, Gelareh Zadeh, S. Suppiah","doi":"10.1093/noajnl/vdae067","DOIUrl":"https://doi.org/10.1093/noajnl/vdae067","url":null,"abstract":"\u0000 Nerve sheath tumors are the most common benign tumors of the spine after meningiomas. They include schwannomas, neurofibroma, and malignant peripheral nerve sheath tumors. These can arise sporadically or in association with tumor predisposition syndromes, including Neurofibromatosis type 1, Neurofibromatosis type 2, and Schwannomatosis. Though surgery is the traditional mainstay of treatment for these tumors, the discovery of the genetic and molecular basis of these diseases in recent decades has prompted investigation into targeted therapies. Here, we give a clinical overview of spinal nerve sheath tumors, their imaging features, current management practices, and explore ongoing advances in systemic therapies.","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141830530","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Dongqing Sun, E. Schaft, Bibi M van Stempvoort, T. Gebbink, Maryse A van 't Klooster, Pieter van Eijsden, Sandra M A van der Salm, J. Dankbaar, M. Zijlmans, Pierre A Robe
{"title":"Intraoperative mapping of epileptogenic foci and tumor infiltration in neuro-oncology patients with epilepsy","authors":"Dongqing Sun, E. Schaft, Bibi M van Stempvoort, T. Gebbink, Maryse A van 't Klooster, Pieter van Eijsden, Sandra M A van der Salm, J. Dankbaar, M. Zijlmans, Pierre A Robe","doi":"10.1093/noajnl/vdae125","DOIUrl":"https://doi.org/10.1093/noajnl/vdae125","url":null,"abstract":"\u0000 \u0000 \u0000 Epileptogenesis and glioma growth have a bidirectional relationship. We hypothesized people with gliomas can benefit from removal of epileptic tissue and that tumor-related epileptic activity may signify tumor infiltration in peritumoral regions. We investigated whether intraoperative electrocorticography (ioECoG) could improve seizure outcome in oncological glioma surgery, and vice versa, what epileptic activity (EA) tells about tumor infiltration.\u0000 \u0000 \u0000 \u0000 We prospectively included patients who underwent (awake) ioECoG-assisted diffuse-glioma resection through the oncological trajectory. IoECoG-tailoring strategy relied on ictal and interictal EA (spikes and sharp-waves). Brain tissue, where EA was recorded, was assigned for histopathological examination separate from the rest of the tumor. Weibull regression was performed to assess how residual EA and extent of resection (EOR) related to the time-to-seizure recurrence, and we investigated which type of EA predicted tumor infiltration.\u0000 \u0000 \u0000 \u0000 Fifty-two patients were included. Residual spikes after resection were associated with seizure recurrence in patients with isocitrate dehydrogenase (IDH) mutant astrocytoma or oligodendroglioma (HR=7.6[1.4–40.0], p-value=0.01), independent from the EOR. This was not observed in IDH-wildtype tumors. All tissue samples resected based on interictal spikes were infiltrated by tumor, even if MRI did not show abnormalities.\u0000 \u0000 \u0000 \u0000 Complete resection of epileptogenic foci in ioECoG may promote seizure control in IDH-mutant gliomas. The cohort size of IDH-wildtype tumors was too limited to draw definitive conclusions. Interictal spikes may indicate tumor infiltration even when this area appears normal on MRI. Integrating electrophysiology guidance into oncological tumor surgery could contribute to improved seizure outcome and precise guidance for radical tumor resection.\u0000","PeriodicalId":94157,"journal":{"name":"Neuro-oncology advances","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2024-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141642577","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}