Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.309
Filter
1.
Cancer Med ; 13(13): e7369, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38970209

ABSTRACT

BACKGROUND: The diagnosis of glioma has advanced since the release of the WHO 2021 classification with more molecular alterations involved in the integrated diagnostic pathways. Our study aimed to present our experience with the clinical features and management of astrocytoma, IDH mutant based on the latest WHO classification. METHODS: Patients diagnosed with astrocytoma, IDH-mutant based on the WHO 5th edition classification of CNS tumors at our center from January 2009 to January 2022 were included. Patients were divided into WHO 2-3 grade group and WHO 4 grade group. Integrate diagnoses were retrospectively confirmed according to WHO 2016 and 2021 classification. Clinical and MRI characteristics were reviewed, and survival analysis was performed. RESULTS: A total of 60 patients were enrolled. 21.67% (13/60) of all patients changed tumor grade from WHO 4th edition classification to WHO 5th edition. Of these, 21.43% (6/28) of grade II astrocytoma and 58.33% (7/12) of grade III astrocytoma according to WHO 4th edition classification changed to grade 4 according to WHO 5th edition classification. Sex (p = 0.042), recurrent glioma (p = 0.006), and Ki-67 index (p < 0.001) of pathological examination were statistically different in the WHO grade 2-3 group (n = 27) and WHO grade 4 group (n = 33). CDK6 (p = 0.004), FGFR2 (p = 0.003), and MYC (p = 0.004) alterations showed an enrichment in the WHO grade 4 group. Patients with higher grade showed shorter mOS (mOS = 75.9 m, 53.6 m, 26.4 m for grade 2, 3, and 4, respectively, p = 0.01). CONCLUSIONS: Patients diagnosed as WHO grade 4 according to the 5th edition WHO classification based on molecular alterations are more likely to have poorer prognosis. Therefore, treatment should be tailored to their individual needs. Further research is needed for the management of IDH-mutant astrocytoma is needed in the future.


Subject(s)
Astrocytoma , Magnetic Resonance Imaging , Mutation , Neoplasm Grading , World Health Organization , Humans , Astrocytoma/genetics , Astrocytoma/classification , Astrocytoma/pathology , Astrocytoma/diagnostic imaging , Male , Female , Retrospective Studies , Middle Aged , Adult , Magnetic Resonance Imaging/methods , Prognosis , Isocitrate Dehydrogenase/genetics , Central Nervous System Neoplasms/classification , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/pathology , Central Nervous System Neoplasms/diagnostic imaging , Aged , Young Adult , Brain Neoplasms/classification , Brain Neoplasms/genetics , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Brain Neoplasms/mortality , Adolescent
2.
Genes (Basel) ; 15(6)2024 May 28.
Article in English | MEDLINE | ID: mdl-38927643

ABSTRACT

Azoospermia is a form of male infertility characterized by a complete lack of spermatozoa in the ejaculate. Sertoli cell-only syndrome (SCOS) is the most severe form of azoospermia, where no germ cells are found in the tubules. Recently, FANCM gene variants were reported as novel genetic causes of spermatogenic failure. At the same time, FANCM variants are known to be associated with cancer predisposition. We performed whole-exome sequencing on a male patient diagnosed with SCOS and a healthy father. Two compound heterozygous missense mutations in the FANCM gene were found in the patient, both being inherited from his parents. After the infertility assessment, the patient was diagnosed with diffuse astrocytoma. Immunohistochemical analyses in the testicular and tumor tissues of the patient and adequate controls showed, for the first time, not only the existence of a cytoplasmic and not nuclear pattern of FANCM in astrocytoma but also in non-mitotic neurons. In the testicular tissue of the SCOS patient, cytoplasmic anti-FANCM staining intensity appeared lower than in the control. Our case report raises a novel possibility that the infertile carriers of FANCM gene missense variants could also be prone to cancer development.


Subject(s)
Astrocytoma , Mutation, Missense , Sertoli Cell-Only Syndrome , Humans , Male , Astrocytoma/genetics , Astrocytoma/pathology , Astrocytoma/diagnosis , Sertoli Cell-Only Syndrome/genetics , Sertoli Cell-Only Syndrome/pathology , Adult , Exome Sequencing , DNA Helicases/genetics , Azoospermia/genetics , Azoospermia/pathology , Azoospermia/diagnosis
3.
Acta Neuropathol Commun ; 12(1): 95, 2024 06 14.
Article in English | MEDLINE | ID: mdl-38877600

ABSTRACT

MYC dysregulation is pivotal in the onset and progression of IDH-mutant gliomas, mostly driven by copy-number alterations, regulatory element alterations, or epigenetic changes. Our pilot analysis uncovered instances of relative MYC overexpression without alterations in the proximal MYC network (PMN), prompting a deeper investigation into potential novel oncogenic mechanisms. Analysing comprehensive genomics profiles of 236 "IDH-mutant 1p/19q non-co-deleted" lower-grade gliomas from The Cancer Genome Atlas, we identified somatic genomic alterations within the PMN. In tumours without PMN-alterations but with MYC-overexpression, genes correlated with MYC-overexpression were identified. Our analyses yielded that 86/236 of astrocytomas exhibited no PMN-alterations, a subset of 21/86 displaying relative MYC overexpression. Within this subset, we discovered 42 genes inversely correlated with relative MYC expression, all on 19q. Further analysis pinpointed a minimal common region at 19q13.43, encompassing 15 genes. The inverse correlations of these 15 genes with relative MYC overexpression were re-confirmed using independent scRNAseq data. Further, the micro-deleted astrocytoma subset displayed significantly higher genomic instability compared to WT cases, but lower instability compared to PMN-hit cases. This newly identified 19q micro-deletion represents a potential novel mechanism underlying MYC dysregulation in astrocytomas. Given the prominence of 19q loss in IDH-mutant gliomas, our findings bear significant implications for understanding gliomagenesis.


Subject(s)
Astrocytoma , Brain Neoplasms , Chromosome Deletion , Chromosomes, Human, Pair 19 , Isocitrate Dehydrogenase , Proto-Oncogene Proteins c-myc , Humans , Isocitrate Dehydrogenase/genetics , Astrocytoma/genetics , Astrocytoma/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Chromosomes, Human, Pair 19/genetics , Mutation
4.
In Vivo ; 38(4): 1671-1676, 2024.
Article in English | MEDLINE | ID: mdl-38936911

ABSTRACT

BACKGROUND/AIM: Gliomas are highly heterogeneous malignancies originating from diverse cell types within the brain. Although their precise etiology is frequently unknown, risk factors, such as chemical exposure, radiation, and specific uncommon genetic disorders have been identified. Diagnosis typically entails imaging tests, such as magnetic resonance imaging and computed tomography, complemented by a biopsy for confirmation, which may be further validated through genetic testing. CASE REPORT: Next-generation sequencing technology revealed germline co-deletion deletion of cyclin-dependent kinase inhibitor 2 A and B genes (CDKN2A and CDKN2B) in a patient diagnosed with pleomorphic xanthoastrocytoma based on the tumor's molecular characteristics. Following this result, we performed focused genetic analysis with use of multiplex ligation-dependent probe amplification technology for the mother that revealed the same co-deletion. Moreover, due to the father's neuroendocrine pancreatic cancer, application of the NGS technology detected a pathogenic variant in the BRCA1-interacting helicase 1 (BRIP1) gene. Comprehensive multi-gene testing conducted within the familial context, marked by a varied spectrum of cancer type, revealed a constellation of genetic predispositions. CONCLUSION: This case study underscores the critical importance of molecular testing for tumor characterization and highlights the pivotal role of genetic testing in facilitating early intervention and screening for at-risk family members. Furthermore, the identification of germline co-deletions in cancer lays the foundation for the development of targeted therapeutic strategies aimed at restoring normal cellular regulation and improving patient management.


Subject(s)
Astrocytoma , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p16 , Germ-Line Mutation , Humans , Cyclin-Dependent Kinase Inhibitor p16/genetics , Astrocytoma/genetics , Astrocytoma/pathology , Cyclin-Dependent Kinase Inhibitor p15/genetics , Germ-Line Mutation/genetics , High-Throughput Nucleotide Sequencing , Genetic Predisposition to Disease , Male , Female , Adult , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Pedigree , Magnetic Resonance Imaging , Gene Deletion
5.
Oncol Res ; 32(6): 1037-1045, 2024.
Article in English | MEDLINE | ID: mdl-38827324

ABSTRACT

Background: The dysregulation of Isocitrate dehydrogenase (IDH) and the subsequent production of 2-Hydroxyglutrate (2HG) may alter the expression of epigenetic proteins in Grade 4 astrocytoma. The interplay mechanism between IDH, O-6-methylguanine-DNA methyltransferase (MGMT)-promoter methylation, and protein methyltransferase proteins-5 (PRMT5) activity, with tumor progression has never been described. Methods: A retrospective cohort of 34 patients with G4 astrocytoma is classified into IDH-mutant and IDH-wildtype tumors. Both groups were tested for MGMT-promoter methylation and PRMT5 through methylation-specific and gene expression PCR analysis. Inter-cohort statistical significance was evaluated. Results: Both IDH-mutant WHO grade 4 astrocytomas (n = 22, 64.7%) and IDH-wildtype glioblastomas (n = 12, 35.3%) had upregulated PRMT5 gene expression except in one case. Out of the 22 IDH-mutant tumors, 10 (45.5%) tumors showed MGMT-promoter methylation and 12 (54.5%) tumors had unmethylated MGMT. All IDH-wildtype tumors had unmethylated MGMT. There was a statistically significant relationship between MGMT-promoter methylation and IDH in G4 astrocytoma (p-value = 0.006). Statistically significant differences in progression-free survival (PFS) were also observed among all G4 astrocytomas that expressed PRMT5 and received either temozolomide (TMZ) or TMZ plus other chemotherapies, regardless of their IDH or MGMT-methylation status (p-value=0.0014). Specifically, IDH-mutant tumors that had upregulated PRMT5 activity and MGMT-promoter methylation, who received only TMZ, have exhibited longer PFS. Conclusions: The relationship between PRMT5, MGMT-promoter, and IDH is not tri-directional. However, accumulation of D2-hydroxyglutarate (2-HG), which partially activates 2-OG-dependent deoxygenase, may not affect their activities. In IDH-wildtype glioblastomas, the 2HG-2OG pathway is typically inactive, leading to PRMT5 upregulation. TMZ alone, compared to TMZ-plus, can increase PFS in upregulated PRMT5 tumors. Thus, using a PRMT5 inhibitor in G4 astrocytomas may help in tumor regression.


Subject(s)
Astrocytoma , DNA Methylation , DNA Modification Methylases , DNA Repair Enzymes , Disease Progression , Isocitrate Dehydrogenase , Mutation , Promoter Regions, Genetic , Protein-Arginine N-Methyltransferases , Tumor Suppressor Proteins , Humans , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , Isocitrate Dehydrogenase/genetics , Male , Female , Astrocytoma/genetics , Astrocytoma/pathology , Middle Aged , Adult , Retrospective Studies , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Neoplasm Grading , Aged , Temozolomide/therapeutic use , Temozolomide/pharmacology , Gene Expression Regulation, Neoplastic
7.
ESMO Open ; 9(6): 103485, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38833969

ABSTRACT

BACKGROUND: The World Health Organization (WHO) 2021 classification of central nervous system (CNS) tumors classified astrocytoma isocitrate dehydrogenase-mutant (A IDHm) with either microvascular proliferation and/or necrosis or homozygous deletion of CDKN2A/B as CNS grade 4 (CNS WHO G4), introducing a distinct entity and posing new challenges to physicians for appropriate management and prognostication. PATIENTS AND METHODS: We retrospectively collected information about patients diagnosed with A IDHm CNS WHO G4 at three reference neuro-oncological Italian centers and correlated them with survival. RESULTS: A total of 133 patients were included. Patients were young (median age 41 years) and most received post-operative treatment including chemo-radiation (n = 101) and/or temozolomide maintenance (n = 112). With a median follow-up of 51 months, the median overall survival (mOS) was 31.2 months, with a 5-year survival probability of 26%. In the univariate analysis, complete resection (mOS: 40.2 versus 26.3 months, P = 0.03), methyl-guaninemethyltransferase (MGMT) promoter methylation (mOS: 40.7 versus 18 months, P = 0.0136), and absence of telomerase reverse transcriptase (TERT) promoter mutation (mOS: 40.7 versus 18 months, P = 0.0003) correlated with better prognosis. In the multivariate models, lack of TERT promoter mutation [hazard ratio (HR) 0.23, 95% confidence interval (CI) 0.07-0.82, P = 0.024] and MGMT methylation (HR 0.40, 95% CI 0.20-0.81, P = 0.01) remained associated with improved survival. CONCLUSIONS: This is the largest experience in Western countries exploring the prognostic signature of patients with A IDHm CNS G4. Our results show that MGMT promoter methylation and TERT promoter mutation may impact clinical outcomes. This may support physicians in prognostication, clinical management, and design of future studies of this distinct diagnostic entity.


Subject(s)
Astrocytoma , Isocitrate Dehydrogenase , Mutation , Humans , Retrospective Studies , Isocitrate Dehydrogenase/genetics , Astrocytoma/genetics , Astrocytoma/mortality , Astrocytoma/therapy , Male , Female , Adult , Prognosis , Middle Aged , Young Adult , Brain Neoplasms/genetics , DNA Repair Enzymes/genetics , DNA Modification Methylases/genetics , Aged , Telomerase/genetics , Adolescent , Neoplasm Grading , DNA Methylation , Tumor Suppressor Proteins/genetics
8.
Anticancer Res ; 44(7): 3005-3011, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38925840

ABSTRACT

BACKGROUND/AIM: Glioblastoma multiforme (GBM) is one of the most lethal types of brain cancer with a median survival of only 12 months due to its aggressiveness and lack of effective treatment options. Astrocytomas and oligodendrogliomas are classified as low-grade gliomas (LGG) and have the potential to progress into secondary GBM. YAP1 and TAZ are transcriptional co-activators of the hippo pathway and play an important role in tumorigenesis by controlling cell proliferation and differentiation. The aim of this study was to analyze whether YAP1 and TAZ influence the survival in patients with astrocytoma and oligodendroglioma. PATIENTS AND METHODS: A total of 22 patient samples of astrocytoma and 11 samples of oligodendroglioma were analyzed using real-time PCR. We utilized open-access data from The Cancer Genome Atlas (TCGA) focusing on "brain lower grade glioma". mRNA expression rates were used to validate our findings on survival analysis. RESULTS: Expression of YAP1 was twice as high in astrocytoma than in oligodendroglioma, whereas there was no difference in TAZ. In oligodendrogliomas, the expression of TAZ was higher in relapsed than in primary tumors. Patients with astrocytoma having a high YAP1 expression had a significantly shorter overall survival than patients with lower expression (median survival 161 vs. 86 months, p=0.0248). These findings were validated with survival analysis of TCGA data. CONCLUSION: High YAP1 expression shows a high correlation with poorer overall survival in LGG. YAP1 has higher levels of expression in astrocytomas than in oligodendrogliomas.


Subject(s)
Adaptor Proteins, Signal Transducing , Astrocytoma , Brain Neoplasms , Transcription Factors , YAP-Signaling Proteins , Humans , YAP-Signaling Proteins/metabolism , Astrocytoma/metabolism , Astrocytoma/genetics , Astrocytoma/pathology , Astrocytoma/mortality , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Female , Male , Transcription Factors/genetics , Transcription Factors/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Middle Aged , Adult , Neoplasm Grading , Oligodendroglioma/genetics , Oligodendroglioma/metabolism , Oligodendroglioma/pathology , Oligodendroglioma/mortality , Phosphoproteins/metabolism , Phosphoproteins/genetics , Aged , Prognosis , Gene Expression Regulation, Neoplastic , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Trans-Activators/genetics , Trans-Activators/metabolism , Young Adult
9.
Ann Diagn Pathol ; 72: 152318, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38733671

ABSTRACT

BACKGROUND: Pilocytic astrocytoma (PAs) represents a significant portion of childhood primary brain tumors, with distinct histological and radiological features. The prevalence of KIAA1549::BRAF fusion in PAs has been well-established, this study aims to assess the prevalence of KIAA1549::BRAF fusions and explore their associations with tumor characteristics, radiological findings, and patient outcomes in PAs. METHODS: Histologically confirmed cases of PAs from a 5-year period were included in the study. Demographic, histopathological, and radiological data were collected, and immunohistochemistry was performed to characterize tumor markers. FISH and qRT-PCR assays were employed to detect KIAA1549::BRAF fusions. Statistical analyses were conducted to examine associations between fusion status and various other parameters. RESULTS: Histological analysis revealed no significant differences in tumor features based on fusion status. However, younger age groups showed higher fusion prevalence. Radiologically, fusion-positive cases were distributed across different tumor subtypes SE, CWE and NCWE. Survival analysis did not demonstrate a significant impact of fusion status on overall survival, however most cases with recurrence and death harboured KIAA1549::BRAF fusion. Of 200 PAs, KIAA1549::BRAF fusions were detected in 64 % and 74 % of cases via qRT-PCR and FISH, respectively. Concordance between the two platforms was substantial (86 %). CONCLUSION: KIAA1549::BRAF fusions are prevalent in PAs and can be reliably detected using both FISH and qRT-PCR assays. Cost considerations suggest qRT-PCR as a more economical option for fusion detection in routine clinical practice.


Subject(s)
Astrocytoma , Biomarkers, Tumor , Brain Neoplasms , Oncogene Proteins, Fusion , Proto-Oncogene Proteins B-raf , Humans , Female , Male , Child , Astrocytoma/genetics , Astrocytoma/pathology , Child, Preschool , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins B-raf/genetics , Adolescent , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Infant , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Immunohistochemistry/methods , In Situ Hybridization, Fluorescence/methods , Young Adult
11.
AJNR Am J Neuroradiol ; 45(6): 747-752, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38724203

ABSTRACT

BACKGROUND AND PURPOSE: T2-FLAIR mismatch is a highly specific imaging biomarker of IDH-mutant diffuse astrocytoma in adults. It has however also been described in MYB/MYBL1-altered low grade tumors. Our aim was to assess the diagnostic power of the T2-FLAIR mismatch in IDH-mutant astrocytoma and MYB/MYBL1-altered low-grade tumors in children and correlate this mismatch with histology. MATERIALS AND METHODS: We evaluated MR imaging examinations of all pediatric patients, performed at the Princess Máxima Center for Pediatric Oncology and the University Medical Center Utrecht between January 2012 and January 2023, with the histomolecular diagnosis of IDH-mutant astrocytoma, diffuse astrocytoma MYB/MYBL1-altered, or angiocentric glioma, and the presence of T2-FLAIR mismatch was assessed. Histologically, the presence of microcysts in the tumor (a phenomenon suggested to be correlated with T2-FLAIR mismatch in IDH-mutant astrocytomas in adults) was evaluated. RESULTS: Nineteen pediatric patients were diagnosed with either IDH-mutant astrocytoma (n = 8) or MYB/MYBL1-altered tumor (n = 11: diffuse astrocytoma, MYB- or MYBL1-altered n = 8; or angiocentric glioma n = 3). T2-FLAIR mismatch was present in 11 patients, 3 (38%) in the IDH-mutant group and 8 (73%) in the MYB/MYBL1 group. No correlation was found between T2-FLAIR mismatch and the presence of microcysts or an enlarged intercellular space in either IDH-mutant astrocytoma (P = .38 and P = .56, respectively) or MYB/MYBL1-altered tumors (P = .36 and P = .90, respectively). CONCLUSIONS: In our pediatric population, T2-FLAIR mismatch was more often found in MYB/MYBL1-altered tumors than in IDH-mutant astrocytomas. In contrast to what has been reported for IDH-mutant astrocytomas in adults, no correlation was found with microcystic changes in the tumor tissue. This finding challenges the hypothesis that such microcystic changes and/or enlarged intercellular spaces in the tissue of these tumors are an important part of explaining the occurrence of the T2-FLAIR mismatch.


Subject(s)
Astrocytoma , Brain Neoplasms , Glioma , Magnetic Resonance Imaging , Proto-Oncogene Proteins c-myb , Humans , Astrocytoma/diagnostic imaging , Astrocytoma/genetics , Astrocytoma/pathology , Child , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Male , Female , Adolescent , Child, Preschool , Glioma/diagnostic imaging , Glioma/genetics , Glioma/pathology , Magnetic Resonance Imaging/methods , Proto-Oncogene Proteins c-myb/genetics , Trans-Activators/genetics , Biomarkers, Tumor/genetics , Isocitrate Dehydrogenase/genetics , Infant , Mutation , Retrospective Studies , Proto-Oncogene Proteins
12.
Neurosciences (Riyadh) ; 29(2): 139-143, 2024 May.
Article in English | MEDLINE | ID: mdl-38740392

ABSTRACT

Subependymal giant cell astrocytoma (SEGA) is a rare circumscribed astrocytic glioma that occurs in approximately 25% of all tuberous sclerosis (TSC) cases. Herein, we discuss an atypical presentation of SEGA, including the genetic alterations, impact on clinical presentation, and the determinants of each medical and surgical treatment option. A 14-year-old girl presented with intermittent headache and a right intraventricular mass originating near the foramen of Monro. The tumor's proximity to critical structures necessitated maximum safe resection, which improved her symptoms. Histological findings indicated SEGA, and genetic sequencing revealed a TSC2 mutation. However, complete clinical and radiological evaluations failed to reveal TSC. Two months later, a new subependymal nodule was incidentally found. She had a recurrent left occipital horn lesion and diffuse smooth leptomeningeal enhancement with no spine drop metastases. She was administered everolimus as the tumor was considered unresectable. Subsequent imaging revealed a reduction in both residual and new lesions.


Subject(s)
Astrocytoma , Mutation , Tuberous Sclerosis Complex 2 Protein , Humans , Female , Astrocytoma/genetics , Astrocytoma/diagnostic imaging , Astrocytoma/pathology , Tuberous Sclerosis Complex 2 Protein/genetics , Adolescent , Brain Neoplasms/genetics , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Tuberous Sclerosis/genetics , Tuberous Sclerosis/diagnostic imaging , Tuberous Sclerosis/complications
13.
J Neurooncol ; 168(2): 355-365, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38557927

ABSTRACT

PURPOSE: The proximate localization of MTAP, which encodes methylthioadenosine phosphorylase, and CDKN2A/B on Chromosome 9q21 has allowed the loss of MTAP expression as a surrogate for homozygous deletion of CDKN2A/B. This study aimed to determine whether MTAP status correlates with clinical outcomes and 11C-methionine uptake in astrocytomas with IDH mutations. METHODS: We conducted immunohistochemistry for MTAP in 30 patients with astrocytoma, IDH-mutant who underwent 11C-methionine positron emission tomography scans prior to surgical resection. The tumor-to-normal (T/N) ratio of 11C-methionine uptake was calculated using the mean standardized uptake value (SUV) for tumor and normal brain tissues. Cox regression analysis was used for multivariate survival analysis. RESULTS: Among IDH-mutant astrocytomas, 26.7% (8/30) exhibited the loss of cytoplasmic MTAP expression, whereas 73.3% (22/30) tumors retained MTAP expression. The median progression-free survival (PFS) was significantly shorter in patients with MTAP loss than those with MTAP retention (1.88 years vs. 6.80 years, p = 0.003). The median overall survival (OS) was also shorter in patients with MTAP loss than in MTAP-retaining counterparts (5.23 years vs. 10.69 years, p = 0.019). Multivariate analysis identified MTAP status (hazard ratio (HR), 0.081) and extent of resection (HR, 0.104) as independent prognostic factors for PFS. Astrocytomas lacking cytoplasmic MTAP expression showed a significantly higher median T/N ratio for 11C-methionine uptake than tumors retaining MTAP (2.12 vs. 1.65, p = 0.012). CONCLUSION: Our study revealed that the loss of MTAP expression correlates with poor prognosis and an elevated T/N ratio of 11C-methionine uptake in astrocytoma, IDH-mutant.


Subject(s)
Astrocytoma , Brain Neoplasms , Isocitrate Dehydrogenase , Methionine , Mutation , Purine-Nucleoside Phosphorylase , Humans , Purine-Nucleoside Phosphorylase/metabolism , Purine-Nucleoside Phosphorylase/genetics , Astrocytoma/genetics , Astrocytoma/metabolism , Astrocytoma/diagnostic imaging , Astrocytoma/pathology , Astrocytoma/mortality , Female , Male , Methionine/metabolism , Middle Aged , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Brain Neoplasms/mortality , Prognosis , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Adult , Aged , Positron-Emission Tomography , Carbon Radioisotopes , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Young Adult
14.
Acta Radiol ; 65(5): 489-498, 2024 May.
Article in English | MEDLINE | ID: mdl-38644751

ABSTRACT

BACKGROUND: The grading of adult isocitrate dehydrogenase (IDH)-mutant astrocytomas is a crucial prognostic factor. PURPOSE: To investigate the value of conventional magnetic resonance imaging (MRI) features and apparent diffusion coefficient (ADC) in the grading of adult IDH-mutant astrocytomas, and to analyze the correlation between ADC and the Ki-67 proliferation index. MATERIAL AND METHODS: The clinical and MRI data of 82 patients with adult IDH-mutant astrocytoma who underwent surgical resection and molecular genetic testing with IDH and 1p/19q were retrospectively analyzed. The conventional MRI features, ADCmin, ADCmean, and nADC of the tumors were compared using the Kruskal-Wallis single factor ANOVA and chi-square tests. Receiver operating characteristic (ROC) curves were drawn to evaluate conventional MRI and ADC accuracy in differentiating tumor grades. Pearson correlation analysis was performed to determine the correlation between ADC and the Ki-67 proliferation index. RESULTS: The difference in enhancement, ADCmin, ADCmean, and nADC among WHO grade 2, 3, and 4 tumors was statistically significant (all P <0.05). ADCmin showed the preferable diagnostic accuracy for grading WHO grade 2 and 3 tumors (AUC=0.724, sensitivity=63.4%, specificity=80%, positive predictive value (PPV)=62.0%; negative predictive value (NPV)=82.5%), and distinguishing grade 3 from grade 4 tumors (AUC=0.764, sensitivity=70%, specificity=76.2%, PPV=75.0%, NPV=71.4%). Enhancement + ADC model showed an optimal predictive accuracy (grade 2 vs. 3: AUC = 0.759; grade 3 vs. 4: AUC = 0.799). The Ki-67 proliferation index was negatively correlated with ADCmin, ADCmean, and nADC (all P <0.05), and positively correlated with tumor grade. CONCLUSION: Conventional MRI features and ADC are valuable to predict pathological grading of adult IDH-mutant astrocytomas.


Subject(s)
Astrocytoma , Brain Neoplasms , Diffusion Magnetic Resonance Imaging , Isocitrate Dehydrogenase , Ki-67 Antigen , Neoplasm Grading , Humans , Astrocytoma/diagnostic imaging , Astrocytoma/genetics , Astrocytoma/pathology , Male , Female , Isocitrate Dehydrogenase/genetics , Ki-67 Antigen/metabolism , Adult , Middle Aged , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Retrospective Studies , Diffusion Magnetic Resonance Imaging/methods , Aged , Mutation , Cell Proliferation , Young Adult , Sensitivity and Specificity
15.
Neurology ; 102(10): e209352, 2024 May.
Article in English | MEDLINE | ID: mdl-38684041

ABSTRACT

BACKGROUND AND OBJECTIVES: Patients with IDH1/2-mutant lower-grade glioma have a high frequency of seizures. We aimed to investigate the correlations between seizures and tumor/patient characteristics and the impact of surgery and adjuvant treatments (AT) on seizure control along the disease trajectory. METHODS: We retrospectively included patients with IDH1/2-mutant lower-grade glioma who underwent surgery at the neurosurgery divisions of the University of Turin and Milan and were treated at the Division of Neuro-Oncology of Turin. Inclusion criteria were a diagnosis according to the 2021 WHO Classification and presentation with seizures; exclusion criteria were presence of CDKN2A/B homozygous deletion, intense/ring contrast enhancement on MRI at presentation, and small tissue biopsy. We evaluated seizure freedom for 2 months after surgery, 6 months from starting observation or AT, at recurrence, and for 6 months after treatments of recurrence. RESULTS: We included 150 patients. There were 77 (51%) and 31 (21%) patients with IDH-mutant/1p19q-codeleted grade 2 and 3 oligodendroglioma and 30 (20%) and 12 (8%) with IDH-mutant grade 2 and 3 astrocytoma, respectively. Total resection was accomplished in 68 (45%). Seventy-five patients (50%) received AT while the remaining 75 were observed with MRI. After 6 months after AT, 28 of 29 patients (96.5%) displayed seizure reduction, 5 of 28 (18%) being seizure-free. 66 of 124 patients (53%) had seizures at recurrence. After 6 months after second-line treatments, 60 of 66 patients (91%) had seizure reduction, 11 (17%) being seizure-free. In multivariable analyses, grade 3 histology positively correlated with seizure freedom at 2 months after surgery (OR 3.5, 1.4-8.9, p = 0.008), 6 months after AT (OR 9.0, 1.5-54.9, p = 0.017), and 6 months after treatment of recurrence (OR 4.9, 1.5-16.5, p = 0.009). Adjuvant radiotherapy reduced seizures at recurrence in a univariate analysis (OR 0.14, 0.03-0.7, p = 0.020). Patients with seizure freedom after surgery and AT displayed longer progression-free survival (PFS) (65, 24.5-105, vs 48 months, 32-63.5, p = 0.037). DISCUSSION: This study analyzed seizure control in patients with IDH1/2-mutant lower-grade glioma across multiple time points. Grade 3 correlated with better seizure control throughout the entire disease trajectory, and seizure freedom after surgery and AT correlated with a longer PFS regardless of tumor grade. These results could serve as an external control arm in clinical trials evaluating the efficacy on seizures of antitumor agents in patients with IDH-mutant lower-grade glioma.


Subject(s)
Brain Neoplasms , Glioma , Isocitrate Dehydrogenase , Mutation , Seizures , Humans , Isocitrate Dehydrogenase/genetics , Male , Female , Brain Neoplasms/genetics , Brain Neoplasms/complications , Brain Neoplasms/therapy , Brain Neoplasms/surgery , Brain Neoplasms/diagnostic imaging , Middle Aged , Seizures/genetics , Seizures/etiology , Seizures/therapy , Glioma/genetics , Glioma/therapy , Glioma/complications , Glioma/diagnostic imaging , Retrospective Studies , Adult , Aged , Oligodendroglioma/genetics , Oligodendroglioma/therapy , Oligodendroglioma/complications , Oligodendroglioma/surgery , Oligodendroglioma/pathology , Neoplasm Grading , Astrocytoma/genetics , Astrocytoma/therapy , Astrocytoma/complications , Astrocytoma/surgery , Astrocytoma/diagnostic imaging
17.
Nat Commun ; 15(1): 3634, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38688897

ABSTRACT

Central nervous system (CNS) tumors are the leading cause of pediatric cancer death, and these patients have an increased risk for developing secondary neoplasms. Due to the low prevalence of pediatric CNS tumors, major advances in targeted therapies have been lagging compared to other adult tumors. We collect single nuclei RNA-seq data from 84,700 nuclei of 35 pediatric CNS tumors and three non-tumoral pediatric brain tissues and characterize tumor heterogeneity and transcriptomic alterations. We distinguish cell subpopulations associated with specific tumor types including radial glial cells in ependymomas and oligodendrocyte precursor cells in astrocytomas. In tumors, we observe pathways important in neural stem cell-like populations, a cell type previously associated with therapy resistance. Lastly, we identify transcriptomic alterations among pediatric CNS tumor types compared to non-tumor tissues, while accounting for cell type effects on gene expression. Our results suggest potential tumor type and cell type-specific targets for pediatric CNS tumor treatment. Here we address current gaps in understanding single nuclei gene expression profiles of previously under-investigated tumor types and enhance current knowledge of gene expression profiles of single cells of various pediatric CNS tumors.


Subject(s)
Central Nervous System Neoplasms , Ependymoma , Gene Expression Regulation, Neoplastic , Transcriptome , Humans , Child , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/pathology , Central Nervous System Neoplasms/metabolism , Ependymoma/genetics , Ependymoma/pathology , Ependymoma/metabolism , Child, Preschool , Astrocytoma/genetics , Astrocytoma/pathology , Astrocytoma/metabolism , Gene Expression Profiling/methods , Female , RNA-Seq , Male , Adolescent , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Cell Nucleus/metabolism , Cell Nucleus/genetics
18.
Childs Nerv Syst ; 40(7): 2199-2207, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38578478

ABSTRACT

Subependymal giant cell astrocytoma (SEGA) represents a benign brain tumor occurring in 5-20% of individuals diagnosed with tuberous sclerosis complex (TSC), serving as a major diagnostic criterion. The presence of SEGA in a patient often prompts consideration of TSC as a probable diagnosis, given its unique association with this disorder. Typically, only one additional major criterion or two minor criteria are necessary to fulfill the diagnostic criteria for TSC. However, in rare instances, SEGA may manifest in patients without clinical features of TSC, termed solitary SEGA. The occurrence of solitary SEGA in patients lacking both clinical manifestations of TSC and genetic confirmation is extremely rare. Furthermore, the presentation of SEGA with intratumoral bleeding is exceedingly uncommon. Here, we presented a case of bleeding solitary SEGA in non-TSC adolescent who underwent surgery and has remained free of disease for a minimum of 3 years. Genetic analysis of peripheral blood and tumor tissue yielded negative results for TSC-related mutations. While SEGA occurrence in non-TSC patients is uncommon, it remains one of the possible diagnoses of intraventricular tumors. However, comprehensive genetic and physical evaluations are imperative to confirm the TSC status and guide further investigations and follow-up appropriately.


Subject(s)
Astrocytoma , Adolescent , Humans , Astrocytoma/complications , Astrocytoma/genetics , Astrocytoma/surgery , Brain Neoplasms/genetics , Brain Neoplasms/complications , Tuberous Sclerosis/complications , Tuberous Sclerosis/genetics
19.
Zhonghua Bing Li Xue Za Zhi ; 53(5): 439-445, 2024 May 08.
Article in Chinese | MEDLINE | ID: mdl-38678323

ABSTRACT

Objective: To examine whether immunohistochemistry of methylthioadenosine phosphorylase (MTAP) and p16 could be used to predict the CDKN2A status in various brain tumors. Methods: A total of 118 cases of IDH-mutant astrocytomas, 16 IDH-wildtype glioblastoma, 17 polymorphic xanthoastrocytoma (PXA) and 20 meningiomas diagnosed at Xuanwu Hospital, Capital Medical University, Beijing, China from November 2017 to October 2023 were collected and analyzed. The CDKN2A status was detected by using fluorescence in situ hybridization or next-generation sequencing. Expression of MTAP and p16 proteins was detected with immunohistochemistry. The association of loss of MTAP/p16 expression with CDKN2A homozygous/heterozygous deletion was examined. Results: Among the 118 cases of IDH-mutant astrocytoma, 13 cases showed homozygous deletion of CDKN2A. All of them had no expression of MTAP while 9 cases had no expression of p16. Among the 16 cases of IDH wild-type glioblastoma, 6 cases showed homozygous deletion of CDKN2A. All 6 cases had no expression of MTAP, while 3 of these cases had no expression of p16 expression. Among the 17 PXA cases, 4 cases showed homozygous deletion of CDKN2A, and the expression of MTAP and p16 was also absent in these 4 cases. Among the 20 cases of meningiomas, 4 cases showed homozygous deletion of CDKN2A. Their expression of MTAP and p16 was also absent. Among the four types of brain tumors, MTAP was significantly correlated with CDKN2A homozygous deletion (P<0.05), with a sensitivity of 100%. However, it was only significantly correlated with the loss of heterozygosity (LOH) of CDKN2A in astrocytomas (P<0.001). P16 was associated with CDKN2A homozygous deletion in IDH-mutant astrocytoma and PXA (P<0.001), but not with the LOH of CDKN2A. Its sensitivity and specificity were lower than that of MTAP. Conclusions: MTAP could serve as a predictive surrogate for CDKN2A homozygous deletion in adult IDH-mutant astrocytoma, PXA, adult IDH-wildtype glioblastoma and meningioma. However, p16 could only be used in the first two tumor types, and its specificity and sensitivity are lower than that of MTAP.


Subject(s)
Biomarkers, Tumor , Brain Neoplasms , Cyclin-Dependent Kinase Inhibitor p16 , Homozygote , Purine-Nucleoside Phosphorylase , Humans , Purine-Nucleoside Phosphorylase/genetics , Purine-Nucleoside Phosphorylase/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Astrocytoma/genetics , Astrocytoma/metabolism , Meningioma/genetics , Meningioma/metabolism , Meningioma/pathology , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Immunohistochemistry , In Situ Hybridization, Fluorescence , Gene Deletion , Meningeal Neoplasms/genetics , Meningeal Neoplasms/metabolism , Mutation , Male , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Female , Adult , High-Throughput Nucleotide Sequencing
SELECTION OF CITATIONS
SEARCH DETAIL
...