Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Surg Neurol Int ; 15: 286, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39246784

RESUMEN

Background: The objective was to study the effectiveness and diagnostic outcome of frame-based stereotactic brain biopsy (STB) done for contrast non-enhancing lesions using indirect evidence of target selection observed in a plain computed tomography (CT) scan of the head. Methods: Data of patients with contrast non-enhancing brain lesions who underwent STB are collected retrospectively from NIMHANS Bangalore, hospital neurosurgery database from January 2021 to March 2023. Those cases subjected to plain CT scans after fixing the stereotactic frame to the head were included in the study. A final histopathological report analysis of these cases was done to assess the diagnostic accuracy. Results: A total of 27 such cases were biopsied. The mean age of subjects was 44.04 ± 17.812 years. Most subjects were in the age group 31-40 years (29.6%). About 55.6% were male and 44.4% were female. The most common site of biopsy was the frontal lobe. The most common indirect evidence on CT was perilesional edema at 33.3% and periventricular location at 33.3%, followed by intralesional calcification at 11.1%. Our diagnostic accuracy was 92.59%. The asymptomatic hemorrhage rate was 2%, and an increase in perilesional edema was seen in 2% of cases. Conclusion: Indirect targeting is a safe and intuitive method for biopsy of contrast non-enhancing lesions. Due consideration is to be given to various findings visible in non-contrast CT scans of the head as indirect evidence of target selection while performing frame-based STB of contrast non-enhancing lesions. This method will also be helpful in resource-limited centers, especially in low-income countries.

2.
Acta Neurochir (Wien) ; 166(1): 340, 2024 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-39160266

RESUMEN

The authors present the first reported case of MVNT in the thalamus in a 60-year-old man with a 20-year history of epilepsy and recent progressive neurological decline presented for neurosurgical evaluation for a non-enhancing mass predominantly in the right thalamus presumed to be a low-grade glioma. The tumor was subtotally resected using a left contralateral interhemispheric transcallosal approach. Histological and molecular assessment revealed an MVNT with MAPK pathway-activating mutation. The authors also conducted a systematic review of pathology-proven cases of MVNT to provide an up-to-date overview of the literature on the localization, presenting symptoms, and recurrence of this tumor.


Asunto(s)
Neoplasias Encefálicas , Tálamo , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Encefálicas/cirugía , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/diagnóstico por imagen , Tálamo/patología , Tálamo/cirugía , Tálamo/diagnóstico por imagen , Glioma/cirugía , Glioma/patología , Glioma/diagnóstico por imagen
3.
J Neurooncol ; 169(3): 571-579, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38995493

RESUMEN

PURPOSE: The T2-FLAIR mismatch sign is a highly specific diagnostic imaging biomarker for astrocytoma, IDH-mutant. However, a definitive prognostic imaging biomarker has yet to be identified. This study investigated imaging prognostic markers, specifically analyzing T2-weighted and FLAIR images of this tumor. METHODS: We retrospectively analyzed 31 cases of non-enhancing astrocytoma, IDH-mutant treated at our institution, and 30 cases from The Cancer Genome Atlas (TCGA)/The Cancer Imaging Archive (TCIA). We defined "super T2-FLAIR mismatch sign" as having a significantly strong low signal comparable to cerebrospinal fluid at non-cystic lesions rather than just a pale FLAIR low-signal tumor lesion as in conventional T2-FLAIR mismatch sign. Cysts were defined as having a round or oval shape and were excluded from the criteria for the super T2-FLAIR mismatch sign. We evaluated the presence or absence of the T2-FLAIR mismatch sign and super T2-FLAIR mismatch sign using preoperative MRI and analyzed the progression-free survival (PFS) and overall survival (OS) by log-rank test. RESULTS: The T2-FLAIR mismatch sign was present in 17 cases (55%) in our institution and 9 cases (30%) within the TCGA-LGG dataset without any correlation with PFS or OS. However, the super T2-FLAIR mismatch sign was detected in 8 cases (26%) at our institution and 13 cases (43%) in the TCGA-LGG dataset. At our institution, patients displaying the super T2-FLAIR mismatch sign showed significantly extended PFS (122.7 vs. 35.9 months, p = 0.0491) and OS (not reached vs. 116.7 months, p = 0.0232). Similarly, in the TCGA-LGG dataset, those with the super T2-FLAIR mismatch sign exhibited notably longer OS (not reached vs. 44.0 months, p = 0.0177). CONCLUSION: The super T2-FLAIR mismatch is a promising prognostic imaging biomarker for non-enhancing astrocytoma, IDH-mutant.


Asunto(s)
Astrocitoma , Neoplasias Encefálicas , Isocitrato Deshidrogenasa , Imagen por Resonancia Magnética , Mutación , Humanos , Isocitrato Deshidrogenasa/genética , Masculino , Femenino , Astrocitoma/diagnóstico por imagen , Astrocitoma/genética , Astrocitoma/patología , Estudios Retrospectivos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/patología , Pronóstico , Persona de Mediana Edad , Adulto , Anciano , Biomarcadores de Tumor/genética , Adulto Joven , Estudios de Seguimiento , Tasa de Supervivencia
4.
Biomark Med ; 18(9): 431-439, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39007837

RESUMEN

Leptomeningeal metastasis (LM) is a devastating complication of malignancy. Diagnosis relies on both contrast enhancement on imaging and malignant cells in cerebral spinal fluid cytology. Though early detection and prompt intervention improves survival, the detection of LM is limited by false negatives. A rare brainstem imaging finding uncovered specifically in EGFR mutation-positive lung cancer patients may represent an early sign of LM. This sign demonstrates high signal on T2 fluid-attenuated inversion recovery and diffusion-weighted imaging sequences, but paradoxically lacks correlative contrast enhancement. Here we report a case of a 72-year-old female EGFR-positive lung cancer patient who developed this lesion following treatment with two first-generation EGFR tyrosine kinase inhibitors then showed subsequent response to osimertinib, an irreversible third-generation EGFR tyrosine kinase inhibitor.


A non-enhancing, T2 FLAIR hyperintense, diffusion-restricting brainstem lesion in an EGFR-positive lung cancer patient may represent an early indicator of leptomeningeal metastases.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Receptores ErbB , Neoplasias Pulmonares , Inhibidores de Proteínas Quinasas , Humanos , Femenino , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico por imagen , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Anciano , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/diagnóstico por imagen , Tronco Encefálico/patología , Tronco Encefálico/diagnóstico por imagen , Tronco Encefálico/metabolismo , Compuestos de Anilina/uso terapéutico , Acrilamidas/uso terapéutico , Imagen de Difusión por Resonancia Magnética , Indoles , Pirimidinas
5.
Neurooncol Adv ; 6(1): vdae005, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38616896

RESUMEN

Background: Non-enhancing (NE) infiltrating tumor cells beyond the contrast-enhancing (CE) bulk of tumor are potential propagators of recurrence after gross total resection of high-grade glioma. Methods: We leveraged single-nucleus RNA sequencing on 15 specimens from recurrent high-grade gliomas (n = 5) to compare prospectively identified biopsy specimens acquired from CE and NE regions. Additionally, 24 CE and 22 NE biopsies had immunohistochemical staining to validate RNA findings. Results: Tumor cells in NE regions are enriched in neural progenitor cell-like cellular states, while CE regions are enriched in mesenchymal-like states. NE glioma cells have similar proportions of proliferative and putative glioma stem cells relative to CE regions, without significant differences in % Ki-67 staining. Tumor cells in NE regions exhibit upregulation of genes previously associated with lower grade gliomas. Our findings in recurrent GBM paralleled some of the findings in a re-analysis of a dataset from primary GBM. Cell-, gene-, and pathway-level analyses of the tumor microenvironment in the NE region reveal relative downregulation of tumor-mediated neovascularization and cell-mediated immune response, but increased glioma-to-nonpathological cell interactions. Conclusions: This comprehensive analysis illustrates differing tumor and nontumor landscapes of CE and NE regions in high-grade gliomas, highlighting the NE region as an area harboring likely initiators of recurrence in a pro-tumor microenvironment and identifying possible targets for future design of NE-specific adjuvant therapy. These findings also support the aggressive approach to resection of tumor-bearing NE regions.

6.
Neurooncol Adv ; 6(1): vdae013, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38405203

RESUMEN

Background: The T2-FLAIR mismatch sign (T2FM) has nearly 100% specificity for predicting IDH-mutant and 1p/19q noncodeleted astrocytomas (astrocytomas). However, only 18.2%-56.0% of astrocytomas demonstrate a positive T2FM. Methods must be considered for distinguishing astrocytomas from negative T2FM gliomas. In this study, positive T2FM gliomas were manually distinguished from nonenhancing gliomas, and then a support vector machine (SVM) classification model was used to distinguish astrocytomas from negative T2FM gliomas. Methods: Nonenhancing gliomas (regardless of pathological type or grade) diagnosed between January 2022 and October 2022 (N = 300) and November 2022 and March 2023 (N = 196) will comprise the training and validation sets, respectively. Our method for distinguishing astrocytomas from nonenhancing gliomas was examined and validated using the training set and validation set. Results: The specificity of T2FM for predicting astrocytomas was 100% in both the training and validation sets, while the sensitivity was 42.75% and 67.22%, respectively. Using a classification model of SVM based on radiomics features, among negative T2FM gliomas, the accuracy was above 85% when the prediction score was greater than 0.70 in identifying astrocytomas and above 95% when the prediction score was less than 0.30 in identifying nonastrocytomas. Conclusions: Manual screening of positive T2FM gliomas, followed by the SVM classification model to differentiate astrocytomas from negative T2FM gliomas, may be a more effective method for identifying astrocytomas in nonenhancing gliomas.

7.
Neuro Oncol ; 26(1): 115-126, 2024 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-37591790

RESUMEN

BACKGROUND: Given the invasive nature of glioblastoma, tumor cells exist beyond the contrast-enhancing (CE) region targeted during treatment. However, areas of non-enhancing (NE) tumors are difficult to visualize and delineate from edematous tissue. Amine chemical exchange saturation transfer echo planar imaging (CEST-EPI) is a pH-sensitive molecular magnetic resonance imaging technique that was evaluated in its ability to identify infiltrating NE tumors and prognosticate survival. METHODS: In this prospective study, CEST-EPI was obtained in 30 patients and areas with elevated CEST contrast ("CEST+" based on the asymmetry in magnetization transfer ratio: MTRasym at 3 ppm) within NE regions were quantitated. Median MTRasym at 3 ppm and volume of CEST + NE regions were correlated with progression-free survival (PFS). In 20 samples from 14 patients, image-guided biopsies of these areas were obtained to correlate MTRasym at 3 ppm to tumor and non-tumor cell burden using immunohistochemistry. RESULTS: In 15 newly diagnosed and 15 recurrent glioblastoma, higher median MTRasym at 3ppm within CEST + NE regions (P = .007; P = .0326) and higher volumes of CEST + NE tumor (P = .020; P < .001) were associated with decreased PFS. CE recurrence occurred in areas of preoperative CEST + NE regions in 95.4% of patients. MTRasym at 3 ppm was correlated with presence of tumor, cell density, %Ki-67 positivity, and %CD31 positivity (P = .001; P < .001; P < .001; P = .001). CONCLUSIONS: pH-weighted amine CEST-EPI allows for visualization of NE tumor, likely through surrounding acidification of the tumor microenvironment. The magnitude and volume of CEST + NE tumor correlates with tumor cell density, degree of proliferating or "active" tumor, and PFS.


Asunto(s)
Imagen Eco-Planar , Glioblastoma , Humanos , Imagen Eco-Planar/métodos , Glioblastoma/patología , Aminas/química , Estudios Prospectivos , Imagen por Resonancia Magnética/métodos , Concentración de Iones de Hidrógeno , Microambiente Tumoral
8.
Cancers (Basel) ; 15(9)2023 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-37173969

RESUMEN

The preoperative grading of non-enhancing glioma (NEG) remains challenging. Herein, we analyzed clinical and magnetic resonance imaging (MRI) features to predict malignancy in NEG according to the 2021 WHO classification and developed a clinical score, facilitating risk estimation. A discovery cohort (2012-2017, n = 72) was analyzed for MRI and clinical features (T2/FLAIR mismatch sign, subventricular zone (SVZ) involvement, tumor volume, growth rate, age, Pignatti score, and symptoms). Despite a "low-grade" appearance on MRI, 81% of patients were classified as WHO grade 3 or 4. Malignancy was then stratified by: (1) WHO grade (WHO grade 2 vs. WHO grade 3 + 4) and (2) molecular criteria (IDHmut WHO grade 2 + 3 vs. IDHwt glioblastoma + IDHmut astrocytoma WHO grade 4). Age, Pignatti score, SVZ involvement, and T2/FLAIR mismatch sign predicted malignancy only when considering molecular criteria, including IDH mutation and CDKN2A/B deletion status. A multivariate regression confirmed age and T2/FLAIR mismatch sign as independent predictors (p = 0.0009; p = 0.011). A "risk estimation in non-enhancing glioma" (RENEG) score was derived and tested in a validation cohort (2018-2019, n = 40), yielding a higher predictive value than the Pignatti score or the T2/FLAIR mismatch sign (AUC of receiver operating characteristics = 0.89). The prevalence of malignant glioma was high in this series of NEGs, supporting an upfront diagnosis and treatment approach. A clinical score with robust test performance was developed that identifies patients at risk for malignancy.

9.
Magn Reson Imaging ; 100: 18-25, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36924806

RESUMEN

BACKGROUND: Glioblastomas are highly infiltrative tumors, and differentiating between non-enhancing tumors (NETs) and vasogenic edema (Edemas) occurring in the non-enhancing T2-weighted hyperintense area is challenging. Here, we differentiated between NETs and Edemas in glioblastomas using neurite orientation dispersion and density imaging (NODDI) and diffusion tensor imaging (DTI). MATERIALS AND METHODS: Data were collected retrospectively from 21 patients with primary glioblastomas, three with metastasis, and two with meningioma as controls. MRI data included T2 weighted images and contrast enhanced T1 weighted images, NODDI, and DTI. Three neurosurgeons manually assigned volumes of interest (VOIs) to the NETs and Edemas. The DTI and NODDI-derived parameters calculated for each VOI were fractional anisotropy (FA), apparent diffusion coefficient (ADC), intracellular volume fraction (ICVF), isotropic volume fraction (ISOVF), and orientation dispersion index. RESULTS: Sixteen and 14 VOIs were placed on NETs and Edemas, respectively. The ICVF, ISOVF, FA, and ADC values of NETs and Edemas differed significantly (p < 0.01). Receiver operating characteristic curve analysis revealed that using all parameters allowed for improved differentiation of NETs from Edemas (area under the curve = 0.918) from the use of NODDI parameters (0.910) or DTI parameters (0.899). Multiple logistic regression was performed with all parameters, and a predictive formula to differentiate between NETs and Edemas could be created and applied to the edematous regions of the negative control-group images; the tumor prediction degree was well below 0.5, confirming differentiation as edema. CONCLUSIONS: Using NODDI and DTI may prove useful in differentiating NETs from Edemas in the non-contrast T2 hyperintensity region of glioblastomas.


Asunto(s)
Glioblastoma , Neoplasias Meníngeas , Humanos , Imagen de Difusión Tensora/métodos , Glioblastoma/diagnóstico por imagen , Neuritas , Estudios Retrospectivos , Imagen de Difusión por Resonancia Magnética/métodos , Edema
10.
Jpn J Radiol ; 41(3): 335-346, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36342645

RESUMEN

PURPOSE: To investigate the feasibility and dosimetric characteristics of dose painting for non-enhancing low-grade gliomas (NE-LGGs) guided by three-dimensional arterial spin labeling (3D-ASL). MATERIALS AND METHODS: Eighteen patients with NE-LGGs were enrolled. 3D-ASL, T2 fluid-attenuated inversion recovery (T2 Flair) and contrast-enhanced T1-weighted magnetic resonance images were obtained. The gross tumor volume (GTV) was delineated on the T2 Flair. The hyper-perfusion region of the GTV (GTV-ASL) was determined by 3D-ASL, and the GTV-SUB was obtained by subtracting the GTV-ASL from the GTV. The clinical target volume (CTV) was created by iso-tropically expanding the GTV by 1 cm. The planning target volume (PTV), PTV-ASL were obtained by expanding the external margins of the CTV, GTV-ASL, respectively. PTV-SUB was generated by subtracting PTV-ASL from PTV. Three plans were generated for each patient: a conventional plan (plan 1) without dose escalation delivering 95-110% of 45-60 Gy in 1.8-2 Gy fractions to the PTV and two dose-painting plans (plan 2 and plan 3) with dose escalating by 10-20% (range, 50-72 Gy) to the PTV-ASL based on plan 1. The plan 3 was obtained from plan 2 without the maximum dose constraint. The dosimetric differences among the three plans were compared. RESULTS: The volume ratio of the PTV-ASL to the PTV was (23.49 ± 11.94)% (Z = - 3.724, P = 0.000). Compared with plan 1, D2%, D98% and Dmean of PTV-ASL increased by 14.67%,16.17% and 14.31% in plan2 and 19.84%,15.52% and 14.27% in plan3, respectively (P < 0.05); the D2% of the PTV and PTV-SUB increased by 11.89% and 8.34% in plan 2, 15.89% and 8.49% in plan 3, respectively (P < 0.05). The PTV coverages were comparable among the three plans (P > 0.05). In plan 2 and plan 3, the conformity indexes decreased by 18.60% and 12.79%; while the homogeneity index increased by 1.43 and 2 times (P < 0.05). Compared with plan 1, the D0.1 cc of brain stem and Dmax of optic chiasma were slightly increased in plan 2 and plan 3, and the absolute doses met the dose constraint. The doses of the other organs at risk (OARs) were similar among the three plans (P > 0.05). CONCLUSION: The dose delivered to hyper-perfusion volume derived from 3D-ASL can increased by 10-20% while respecting the constraints to the OARs for NE-LGGs, which provides a basis for future individualized and precise radiotherapy, especially if the contrast agent cannot be injected or when contrast enhancement is uncertain.


Asunto(s)
Glioma , Radioterapia de Intensidad Modulada , Humanos , Dosificación Radioterapéutica , Planificación de la Radioterapia Asistida por Computador/métodos , Glioma/diagnóstico por imagen , Glioma/radioterapia , Radioterapia de Intensidad Modulada/métodos , Órganos en Riesgo
11.
J Neurooncol ; 162(3): 481-488, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36577872

RESUMEN

PET imaging using radiolabeled amino acids in addition to MRI has become a valuable diagnostic tool in the clinical management of patients with brain tumors. This review provides a comprehensive overview of PET studies in glioma patients with a mutation in the isocitrate dehydrogenase gene (IDH). A considerable fraction of these tumors typically show no contrast enhancement on MRI, especially when classified as grade 2 according to the World Health Organization classification of Central Nervous System tumors. Major diagnostic challenges in this situation are differential diagnosis, target definition for diagnostic biopsies, delineation of glioma extent for treatment planning, differentiation of treatment-related changes from tumor progression, and the evaluation of response to alkylating agents. The main focus of this review is the role of amino acid PET in this setting. Furthermore, in light of clinical trials using IDH inhibitors targeting the mutated IDH enzyme for treating patients with IDH-mutant gliomas, we also aim to give an outlook on PET probes specifically targeting the IDH mutation, which appear potentially helpful for response assessment.


Asunto(s)
Neoplasias Encefálicas , Glioma , Humanos , Isocitrato Deshidrogenasa/genética , Glioma/diagnóstico por imagen , Glioma/genética , Glioma/terapia , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Tomografía de Emisión de Positrones , Mutación , Aminoácidos/genética
12.
Cureus ; 14(11): e31531, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36532940

RESUMEN

Background and purpose Gadolinium-based contrast agents (GBCAs) have been administered clinically since 1988. They are remarkably well tolerated by children and result in dose-dependent tissue deposition, even in patients with normal renal function. No adverse effects of gadolinium deposition in patients with normal renal function have been established. Given the uncertain effects of gadolinium deposition, we sought to analyze gadolinium use in the imaging follow-up of nonenhancing primary brain neoplasms in children. Materials and methods This retrospective, institutional review board-approved and Health Insurance Portability and Accountability Act-compliant study evaluated pediatric patients who received GBCA in the routine evaluation of brain neoplasms. This special subset included 30 patients (<18 years old) with initially nonenhancing primary intracranial neoplasms who received treatment and follow-up at our institution. Patient data included sex, age from diagnosis to most recent imaging follow-up, number of contrast-enhanced magnetic resonance imaging (MRI) follow-up exams, and histopathology from a biopsy or resection. Results The group had an expected variety of tumors, including low-grade astrocytomas, dysembryoplastic neuroepithelial tumors, oligodendrogliomas, and teratomas. Half of our patients had tumors of unknown histopathology that were not biopsied or resected. The median age at diagnosis was 8.9 years, the median of four follow-up MRIs per patient, and the median follow-up time of four years. Only one of the 30 patients developed an enhancing focus on follow-up MRI that remained stable and asymptomatic over two years and did not require surgical intervention. Conclusion Judicious use of GBCA in children, especially when numerous exams over many years are anticipated, is advised given the data regarding soft-tissue deposition. Preliminary results suggest that it may be feasible to omit GBCA from routine follow-ups of initially nonenhancing brain neoplasms.

13.
Front Oncol ; 12: 866622, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35936687

RESUMEN

Background: Metastatic brain tumors are typically located at the cerebral hemispheres or the cerebellum and most frequently originate from primary breast or lung tumors. Metastatic lesions are usually associated with blood-brain barrier disruption, solid or ring-like contrast enhancement, and perilesional vasogenic edema on brain imaging. Even in cases where metastases are predominantly cystic, enhancement of the minor solid component can be detected. In contrast, non-enhancing secondary brain tumors were only reported in a patient after antiangiogenic treatment with bevacizumab. Case report: We report a case of a 54-year-old male who presented with left-sided weakness and multiple seizures. Brain magnetic resonance imaging revealed a T2-weighted heterogeneous solid tumor in the right frontoparietal parasagittal region, with no apparent enhancement on T1-weighted post-contrast images and no evident perilesional edema. Further MRS analysis revealed markedly increased choline and lipid peaks. The patient underwent craniotomy for tumor removal. Histopathology revealed findings consistent with metastatic non-microcellular neuroendocrine lung cancer. positron emission tomography/computed tomography (PET/CT) revealed a stellate lesion within the right upper lung lobe, compatible with primary lung cancer. Conclusion: Non-enhancing brain metastatic tumors are rarely reported in the literature, usually following antiangiogenic treatment. Here, we report the first ever case of a non-enhancing metastatic brain tumor with no prior history of antiangiogenic treatment, with particular emphasis on the importance of MRS analysis in atypical brain lesions.

14.
NMC Case Rep J ; 9: 199-208, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35974956

RESUMEN

Despite recent signs of progress in diagnostic radiology, it is quite rare that a glioblastoma (GBM) is detected asymptomatically. We describe two patients with asymptomatic nonenhancing GBMs that were not diagnosed with neoplasia at first. The patients had brain scans as medical checkups, and incidentally lesions were detected. In both cases, surgical specimens histopathologically showed no evidence of neoplasia, whereas molecular genetic findings were isocitrate dehydrogenase (IDH)-wildtype, O6-methylguanine-DNA methyltransferase promoter (pMGMT) unmethylated, and telomerase reverse transcriptase (TERT) promoter mutated, which matched to GBM. One patient was observed without adjuvant therapy and the tumor recurred 7 months later. Reoperation was performed, and histopathologically GBM was confirmed with the same molecular diagnosis as the first surgical specimen. Another patient was carefully observed, and chemoradiotherapy was begun 6 months after the operation following the extension of the lesion. Eventually, because of disease progression, both patients deceased. We postulate that in each case, the tumor was not lower-grade glioma but corresponded to the early growth phase of GBM cells. Thus far, cases of malignant transformation from lower-grade glioma or asymptomatic GBM with typical histologic features are reported. Nevertheless, to the best of our knowledge, no such case of nonenhancing, nonhistologically confirmed GBM was reported. We conjecture these cases shed light on the yet unknown natural history of GBM. GBM can take the form of radiological nonenhancing and histological nonneoplastic fashion before typical morphology. Molecular genetic analysis can diagnose atypical preceding GBM, and we recommend early surgical removal and adjuvant treatment.

15.
Front Oncol ; 12: 914507, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35860561

RESUMEN

Objective: The present study aimed to evaluate the feasibility of sub-volume segmentation for radiotherapy planning of adult non-enhancing low-grade gliomas (NE-LGGs) guided by three-dimensional arterial spin labeling (3D-ASL). The differences in high- and low-perfusion areas of NE-LGGs were analyzed using multi-sequence magnetic resonance imaging (MRI) radiomics. Methods: Fifteen adult patients with NE-LGGs were included in the study. MR images, including T1-weighted imaging (T1WI), T2 Propeller, T2 fluid-attenuated inversion recovery (T2 Flair), 3D-ASL, and contrast-enhanced T1WI (CE-T1WI), were obtained. The gross tumor volume (GTV) was delineated according to the hyperintensity on T2 Flair. The GTV was divided into high- and low-perfusion areas, namely GTV-ASL and GTV-SUB, respectively, based on the differences in cerebral blood flow (CBF) value. The volumes and CBF values of high- and low-perfusion areas were measured and compared. The least absolute shrinkage and selection operator (LASSO) regression was used to select the optimal features of all MR maps. Receiver operating characteristic (ROC) curve analysis was used to evaluate the diagnostic accuracy of the absolute CBFmean (aCBFmean), relative CBFmean (rCBFmean, normalized by the CBF value of the normal gray matter), and screened features in differentiating high- and low-perfusion areas. Results: Among the enrolled patients, three (20%) patients with NE-LGGs showed focal intra- and post-radiotherapy contrast enhancement within a prior high-perfusion area of 3D-ASL. The volume ratio of the GTV-ASL to the GTV was (37.08% ± 17.88)% (46.26 ± 44.51 vs. 167.46 ± 209.64 cm3, P = 0.000). The CBFmean in the high-perfusion area was approximately two times of that in the edema area or normal gray matter (66.98 ± 18.03 vs. 35.19 ± 7.75 or 33.92 ± 8.48 ml/100g/min, P = 0.000). Thirteen features were screened, seven of which were extracted from 3D-ASL. The area undercurve (AUC) values of aCBFmean, rCBFmean, and firstorder_10Percentile from 3D-ASL were more than 0.9, of which firstorder_10Percentile was the highest. Their cut-off values were 44.16 ml/100 g/min, 1.49 and 31, respectively. Conclusion: The difference in blood perfusion in the GTV can be quantified and analyzed based on 3D-ASL images for NE-LGGs, which could guide the sub-volume segmentation of the GTV. 3D-ASL should become a routine method for NE-LGGs during simulation and radiotherapy.

16.
Neurooncol Adv ; 4(1): vdac023, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35300151

RESUMEN

Background: Nonenhancing glioma typically have a favorable outcome, but approximately 19-44% have a highly aggressive course due to a glioblastoma genetic profile. The aim of this retrospective study is to use physiological MRI parameters of both perfusion and diffusion to distinguish the molecular profiles of glioma without enhancement at presentation. Methods: Ninety-nine patients with nonenhancing glioma were included, in whom molecular status (including 1p/19q codeletion status and IDH mutation) and preoperative MRI (T2w/FLAIR, dynamic susceptibility-weighted, and diffusion-weighted imaging) were available. Tumors were segmented semiautomatically using ITK-SNAP to derive whole tumor histograms of relative Cerebral Blood Volume (rCBV) and Apparent Diffusion Coefficient (ADC). Tumors were divided into three clinically relevant molecular profiles: IDH mutation (IDHmt) with (n = 40) or without (n = 41) 1p/19q codeletion, and (n = 18) IDH-wildtype (IDHwt). ANOVA, Kruskal-Wallis, and Chi-Square analyses were performed using SPSS. Results: rCBV (mean, median, 75th and 85th percentile) and ADC (mean, median, 15th and 25th percentile) showed significant differences across molecular profiles (P < .01). Posthoc analyses revealed that IDHwt and IDHmt 1p/19q codeleted tumors showed significantly higher rCBV compared to IDHmt 1p/19q intact tumors: mean rCBV (mean, SD) 1.46 (0.59) and 1.35 (0.39) versus 1.08 (0.31), P < .05. Also, IDHwt tumors showed significantly lower ADC compared to IDHmt 1p/19q codeleted and IDHmt 1p/19q intact tumors: mean ADC (mean, SD) 1.13 (0.23) versus 1.27 (0.15) and 1.45 (0.20), P < .001). Conclusions: A combination of low ADC and high rCBV, reflecting high cellularity and high perfusion respectively, separates IDHwt from in particular IDHmt 1p/19q intact glioma.

17.
J Pak Med Assoc ; 72(2): 377-379, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35320200

RESUMEN

Intraventricular tumours are relatively uncommon among all brain tumours, and non-enhancing lesions, mostly subependymoma, are even less frequently reported. Select cases of subependymoma can show variable contrast enhancement as well. Gross total surgical resection is recommended for treating these lesions, with no significant role of adjuvant chemotherapy or radiotherapy.


Asunto(s)
Neoplasias Encefálicas , Neoplasias del Ventrículo Cerebral , Adulto , Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/terapia , Neoplasias del Ventrículo Cerebral/diagnóstico por imagen , Neoplasias del Ventrículo Cerebral/patología , Neoplasias del Ventrículo Cerebral/cirugía , Humanos
18.
Brain Sci ; 12(1)2022 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-35053842

RESUMEN

One of the challenges in glioblastoma (GBM) imaging is to visualize non-enhancing tumor (NET) lesions. The ratio of T1- and T2-weighted images (rT1/T2) is reported as a helpful imaging surrogate of microstructures of the brain. This research study investigated the possibility of using rT1/T2 as a surrogate for the T1- and T2-relaxation time of GBM to visualize NET effectively. The data of thirty-four histologically confirmed GBM patients whose T1-, T2- and contrast-enhanced T1-weighted MRI and 11C-methionine positron emission tomography (Met-PET) were available were collected for analysis. Two of them also underwent MR relaxometry with rT1/T2 reconstructed for all cases. Met-PET was used as ground truth with T2-FLAIR hyperintense lesion, with >1.5 in tumor-to-normal tissue ratio being NET. rT1/T2 values were compared with MR relaxometry and Met-PET. rT1/T2 values significantly correlated with both T1- and T2-relaxation times in a logarithmic manner (p < 0.05 for both cases). The distributions of rT1/T2 from Met-PET high and low T2-FLAIR hyperintense lesions were different and a novel metric named Likeliness of Methionine PET high (LMPH) deriving from rT1/T2 was statistically significant for detecting Met-PET high T2-FLAIR hyperintense lesions (mean AUC = 0.556 ± 0.117; p = 0.01). In conclusion, this research study supported the hypothesis that rT1/T2 could be a promising imaging marker for NET identification.

19.
MAGMA ; 35(1): 63-73, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34994858

RESUMEN

OBJECTIVE: Clinical application of chemical exchange saturation transfer (CEST) can be performed with investigation of amide proton transfer (APT) and nuclear Overhauser enhancement (NOE) effects. Here, we investigated APT- and NOE-weighted imaging based on advanced CEST metrics to map tumor heterogeneity of non-enhancing glioma at 3 T. MATERIALS AND METHODS: APT- and NOE-weighted maps based on Lorentzian difference (LD) and inverse magnetization transfer ratio (MTRREX) were acquired with a 3D snapshot CEST acquisition at 3 T. Saturation power was investigated first by varying B1 (0.5-2 µT) in 5 healthy volunteers then by applying B1 of 0.5 and 1.5 µT in 10 patients with non-enhancing glioma. Tissue contrast (TC) and contrast-to-noise ratios (CNR) were calculated between glioma and normal appearing white matter (NAWM) and grey matter, in APT- and NOE-weighted images. Volume percentages of the tumor showing hypo/hyperintensity (VPhypo/hyper,CEST) in APT/NOE-weighted images were calculated for each patient. RESULTS: LD APT resulting from using a B1 of 1.5 µT was found to provide significant positive TCtumor,NAWM and MTRREX NOE (B1 of 1.5 µT) provided significant negative TCtumor,NAWM in tissue differentiation. MTRREX-based NOE imaging under 1.5 µT provided significantly larger VPhypo,CEST than MTRREX APT under 1.5 µT. CONCLUSION: This work showed that with a rapid CEST acquisition using a B1 saturation power of 1.5 µT and covering the whole tumor, analysis of both LD APT and MTRREX NOE allows for observing tumor heterogeneity, which will be beneficial in future studies using CEST-MRI to improve imaging diagnostics for non-enhancing glioma.


Asunto(s)
Neoplasias Encefálicas , Glioma , Amidas , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/patología , Dimaprit/análogos & derivados , Glioma/diagnóstico por imagen , Glioma/patología , Voluntarios Sanos , Humanos , Imagen por Resonancia Magnética/métodos , Protones
20.
J Neurosurg ; : 1-11, 2022 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-34996044

RESUMEN

OBJECTIVE: Many neurosurgeons resect nonenhancing low-grade gliomas (LGGs) by using an inside-out piecemeal resection (PMR) technique. At the authors' institution they have increasingly used a circumferential, perilesional, sulcus-guided resection (SGR) technique. This technique has not been well described and there are limited data on its effectiveness. The authors describe the SGR technique and assess the extent to which SGR correlates with extent of resection and neurological outcome. METHODS: The authors identified all patients with newly diagnosed LGGs who underwent resection at their institution over a 22-year period. Demographics, presenting symptoms, intraoperative data, method of resection (SGR or PMR), volumetric imaging data, and postoperative outcomes were obtained. Univariate analyses used ANOVA and Fisher's exact test. Multivariate analyses were performed using multivariate logistic regression. RESULTS: Newly diagnosed LGGs were resected in 519 patients, 208 (40%) using an SGR technique and 311 (60%) using a PMR technique. The median extent of resection in the SGR group was 84%, compared with 77% in the PMR group (p = 0.019). In multivariate analysis, SGR was independently associated with a higher rate of complete (100%) resection (27% vs 18%) (OR 1.7, 95% CI 1.1-2.6; p = 0.03). SGR was also associated with a statistical trend toward lower rates of postoperative neurological complications (11% vs 16%, p = 0.09). A subset analysis of tumors located specifically in eloquent brain demonstrated SGR to be as safe as PMR. CONCLUSIONS: The authors describe the SGR technique used to resect LGGs and show that SGR is independently associated with statistically significantly higher rates of complete resection, without an increase in neurological complications, than with PMR. SGR technique should be considered when resecting LGGs.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA