Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
medRxiv ; 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38947051

RESUMEN

Background: Meningioma risk factors include older age, female sex, and African-American race. There are limited data exploring how meningioma risk in African-Americans varies across the lifespan, interacts with sex, and differs by tumor grade. Methods: The Central Brain Tumor Registry of the United States (CBTRUS) is a population-based registry covering the entire U.S. population. Meningioma diagnoses from 2004-2019 were used to calculate incidence rate ratios (IRRs) for non-Hispanic Black individuals (NHB) compared to non-Hispanic white individuals (NHW) across 10-year age intervals, and stratified by sex and by WHO tumor grade. Results: 53,890 NHB individuals and 322,373 NHW individuals with an intracranial meningioma diagnosis were included in analyses. Beginning in young adulthood, the NHB-to-NHW IRR was elevated for both grade 1 and grade 2/3 tumors. The IRR peaked in the seventh decade of life regardless of grade, and was higher for grade 2/3 tumors (IRR=1.57; 95% CI: 1.46-1.69) than grade 1 tumors (IRR=1.27; 95% CI: 1.25-1.30) in this age group. The NHB-to-NHW IRR was elevated in females (IRR=1.17; 95% CI: 1.16-1.18) and further elevated in males (IRR=1.28; 95% CI: 1.26-1.30), revealing synergistic interaction between NHB race/ethnicity and male sex (P Interaction =0.001). Conclusions: Relative to NHW individuals, NHB individuals are at elevated risk of meningioma from young adulthood through old age. NHB race/ethnicity conferred higher risk of meningioma among men than women, and higher risk of developing WHO grade 2/3 tumors. Results identify meningioma as a significant source of racial disparities in neuro-oncology and may help to improve preoperative predictions of meningioma grade.

2.
Radiol Artif Intell ; : e240076, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38984984

RESUMEN

"Just Accepted" papers have undergone full peer review and have been accepted for publication in Radiology: Artificial Intelligence. This article will undergo copyediting, layout, and proof review before it is published in its final version. Please note that during production of the final copyedited article, errors may be discovered which could affect the content. Purpose To develop a deep learning algorithm to predict 2-year neurodevelopmental outcomes in neonates with hypoxic-ischemic encephalopathy (HIE) using MRI and basic clinical data. Materials and Methods In this study, MRI data of term neonates with encephalopathy in the High Dose Erythropoietin for Asphyxia (HEAL) trial (ClinicalTrials.gov: NCT02811263), who were enrolled from 17 institutions between January 25th, 2017 and October ninth, 2019, were retrospectively analyzed. The harmonized MRI protocol included T1-weighted, T2-weighted, and diffusion tensor imaging. Deep learning classifiers were trained to predict the primary outcome of the HEAL trial (death or any neurodevelopmental impairment [NDI] at 2 years) using multisequence MRI and basic clinical variables, including sex and gestational age at birth. Model performance was evaluated on a test sets comprising 10% of cases from 15 institutions (in-distribution test set, n = 41) and 100% of cases from 2 institutions (out-of-distribution test set, n = 41). Model performance in predicting additional secondary outcomes, including death alone, was also assessed. Results For the 414 neonates (mean gestational age, 39 weeks ± 1.4, 232 males, 182 females), in the study cohort, 198 (48%) died or had any NDI at 2 years. The deep learning model achieved an area under the receiver operating characteristic curve (AUC) of 0.74 (95% CI: 0.60-0.86) and 63% accuracy on the in-distribution test set and an AUC of 0.77 (95% CI: 0.63-0.90) and 78% accuracy on the out-of-distribution test set. Performance was similar or better for predicting secondary outcomes. Conclusion Deep learning analysis of neonatal brain MRI yielded high performance for predicting 2-year neurodevelopmental outcomes. ©RSNA, 2024.

5.
Sci Rep ; 14(1): 4583, 2024 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-38403673

RESUMEN

Brain extraction, or skull-stripping, is an essential data preprocessing step for machine learning approaches to brain MRI analysis. Currently, there are limited extraction algorithms for the neonatal brain. We aim to adapt an established deep learning algorithm for the automatic segmentation of neonatal brains from MRI, trained on a large multi-institutional dataset for improved generalizability across image acquisition parameters. Our model, ANUBEX (automated neonatal nnU-Net brain MRI extractor), was designed using nnU-Net and was trained on a subset of participants (N = 433) enrolled in the High-dose Erythropoietin for Asphyxia and Encephalopathy (HEAL) study. We compared the performance of our model to five publicly available models (BET, BSE, CABINET, iBEATv2, ROBEX) across conventional and machine learning methods, tested on two public datasets (NIH and dHCP). We found that our model had a significantly higher Dice score on the aggregate of both data sets and comparable or significantly higher Dice scores on the NIH (low-resolution) and dHCP (high-resolution) datasets independently. ANUBEX performs similarly when trained on sequence-agnostic or motion-degraded MRI, but slightly worse on preterm brains. In conclusion, we created an automatic deep learning-based neonatal brain extraction algorithm that demonstrates accurate performance with both high- and low-resolution MRIs with fast computation time.


Asunto(s)
Imagen por Resonancia Magnética , Neuroimagen , Humanos , Recién Nacido , Encéfalo/diagnóstico por imagen , Cabeza , Procesamiento de Imagen Asistido por Computador/métodos , Imagen por Resonancia Magnética/métodos , Neuroimagen/métodos , Cráneo , Estudios Multicéntricos como Asunto
6.
Neuro Oncol ; 26(6): 1152-1162, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38170451

RESUMEN

BACKGROUND: Laser interstitial thermal therapy (LITT) of intracranial tumors or radiation necrosis enables tissue diagnosis, cytoreduction, and rapid return to systemic therapies. Ablated tissue remains in situ, resulting in characteristic post-LITT edema associated with transient clinical worsening and complicating post-LITT response assessment. METHODS: All patients receiving LITT at a single center for tumors or radiation necrosis from 2015 to 2023 with ≥9 months of MRI follow-up were included. An nnU-Net segmentation model was trained to automatically segment contrast-enhancing lesion volume (CeLV) of LITT-treated lesions on T1-weighted images. Response assessment was performed using volumetric measurements. RESULTS: Three hundred and eighty four unique MRI exams of 61 LITT-treated lesions and 6 control cases of medically managed radiation necrosis were analyzed. Automated segmentation was accurate in 367/384 (95.6%) images. CeLV increased to a median of 68.3% (IQR 35.1-109.2%) from baseline at 1-3 months from LITT (P = 0.0012) and returned to baseline thereafter. Overall survival (OS) for LITT-treated patients was 39.1 (9.2-93.4) months. Lesion expansion above 40% from volumetric nadir or baseline was considered volumetric progression. Twenty-one of 56 (37.5%) patients experienced progression for a volumetric progression-free survival of 21.4 (6.0-93.4) months. Patients with volumetric progression had worse OS (17.3 vs 62.1 months, P = 0.0015). CONCLUSIONS: Post-LITT CeLV expansion is quantifiable and resolves within 6 months of LITT. Development of response assessment criteria for LITT-treated lesions is feasible and should be considered for clinical trials. Automated lesion segmentation could speed the adoption of volumetric response criteria in clinical practice.


Asunto(s)
Neoplasias Encefálicas , Terapia por Láser , Humanos , Femenino , Masculino , Terapia por Láser/métodos , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/diagnóstico por imagen , Persona de Mediana Edad , Imagen por Resonancia Magnética/métodos , Adulto , Redes Neurales de la Computación , Anciano , Estudios de Seguimiento , Estudios Retrospectivos , Pronóstico , Hipertermia Inducida/métodos , Aprendizaje Profundo
7.
ArXiv ; 2024 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-37292481

RESUMEN

Pediatric tumors of the central nervous system are the most common cause of cancer-related death in children. The five-year survival rate for high-grade gliomas in children is less than 20%. Due to their rarity, the diagnosis of these entities is often delayed, their treatment is mainly based on historic treatment concepts, and clinical trials require multi-institutional collaborations. The MICCAI Brain Tumor Segmentation (BraTS) Challenge is a landmark community benchmark event with a successful history of 12 years of resource creation for the segmentation and analysis of adult glioma. Here we present the CBTN-CONNECT-DIPGR-ASNR-MICCAI BraTS-PEDs 2023 challenge, which represents the first BraTS challenge focused on pediatric brain tumors with data acquired across multiple international consortia dedicated to pediatric neuro-oncology and clinical trials. The BraTS-PEDs 2023 challenge focuses on benchmarking the development of volumentric segmentation algorithms for pediatric brain glioma through standardized quantitative performance evaluation metrics utilized across the BraTS 2023 cluster of challenges. Models gaining knowledge from the BraTS-PEDs multi-parametric structural MRI (mpMRI) training data will be evaluated on separate validation and unseen test mpMRI dataof high-grade pediatric glioma. The CBTN-CONNECT-DIPGR-ASNR-MICCAI BraTS-PEDs 2023 challenge brings together clinicians and AI/imaging scientists to lead to faster development of automated segmentation techniques that could benefit clinical trials, and ultimately the care of children with brain tumors.

8.
J Magn Reson Imaging ; 2023 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-37905681

RESUMEN

Gadolinium contrast is an important agent in magnetic resonance imaging (MRI), particularly in neuroimaging where it can help identify blood-brain barrier breakdown from an inflammatory, infectious, or neoplastic process. However, gadolinium contrast has several drawbacks, including nephrogenic systemic fibrosis, gadolinium deposition in the brain and bones, and allergic-like reactions. As computer hardware and technology continues to evolve, machine learning has become a possible solution for eliminating or reducing the dose of gadolinium contrast. This review summarizes the clinical uses of gadolinium contrast, the risks of gadolinium contrast, and state-of-the-art machine learning methods that have been applied to reduce or eliminate gadolinium contrast administration, as well as their current limitations, with a focus on neuroimaging applications. EVIDENCE LEVEL: 3 TECHNICAL EFFICACY: Stage 1.

9.
Radiology ; 308(3): e223262, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37698478

RESUMEN

Background Multiple qualitative scoring systems have been created to capture the imaging severity of hypoxic ischemic brain injury. Purpose To evaluate quantitative volumes of acute brain injury at MRI in neonates with hypoxic ischemic brain injury and correlate these findings with 24-month neurodevelopmental outcomes and qualitative brain injury scoring by radiologists. Materials and Methods In this secondary analysis, brain diffusion-weighted MRI data from neonates in the High-dose Erythropoietin for Asphyxia and Encephalopathy trial, which recruited participants between January 2017 and October 2019, were analyzed. Volume of acute brain injury, defined as brain with apparent diffusion coefficient (ADC) less than 800 × 10-6 mm2/sec, was automatically computed across the whole brain and within the thalami and white matter. Outcomes of death and neurodevelopmental impairment (NDI) were recorded at 24-month follow-up. Associations between the presence and volume (in milliliters) of acute brain injury with 24-month outcomes were evaluated using multiple logistic regression. The correlation between quantitative acute brain injury volume and qualitative MRI scores was assessed using the Kendall tau-b test. Results A total of 416 neonates had available MRI data (mean gestational age, 39.1 weeks ± 1.4 [SD]; 235 male) and 113 (27%) showed evidence of acute brain injury at MRI. Of the 387 participants with 24-month follow-up data, 185 (48%) died or had any NDI. Volume of acute injury greater than 1 mL (odds ratio [OR], 13.9 [95% CI: 5.93, 32.45]; P < .001) and presence of any acute injury in the brain (OR, 4.5 [95% CI: 2.6, 7.8]; P < .001) were associated with increased odds of death or any NDI. Quantitative whole-brain acute injury volume was strongly associated with radiologists' qualitative scoring of diffusion-weighted images (Kendall tau-b = 0.56; P < .001). Conclusion Automated quantitative volume of brain injury is associated with death, moderate to severe NDI, and cerebral palsy in neonates with hypoxic ischemic encephalopathy and correlated well with qualitative MRI scoring of acute brain injury. Clinical trial registration no. NCT02811263 © RSNA, 2023 Supplemental material is available for this article. See also the editorial by Huisman in this issue.


Asunto(s)
Lesiones Encefálicas , Hipoxia-Isquemia Encefálica , Recién Nacido , Masculino , Humanos , Lactante , Benchmarking , Imagen por Resonancia Magnética , Imagen de Difusión por Resonancia Magnética , Encéfalo/diagnóstico por imagen , Hipoxia-Isquemia Encefálica/diagnóstico por imagen
10.
ArXiv ; 2023 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-37608932

RESUMEN

Automated brain tumor segmentation methods have become well-established and reached performance levels offering clear clinical utility. These methods typically rely on four input magnetic resonance imaging (MRI) modalities: T1-weighted images with and without contrast enhancement, T2-weighted images, and FLAIR images. However, some sequences are often missing in clinical practice due to time constraints or image artifacts, such as patient motion. Consequently, the ability to substitute missing modalities and gain segmentation performance is highly desirable and necessary for the broader adoption of these algorithms in the clinical routine. In this work, we present the establishment of the Brain MR Image Synthesis Benchmark (BraSyn) in conjunction with the Medical Image Computing and Computer-Assisted Intervention (MICCAI) 2023. The primary objective of this challenge is to evaluate image synthesis methods that can realistically generate missing MRI modalities when multiple available images are provided. The ultimate aim is to facilitate automated brain tumor segmentation pipelines. The image dataset used in the benchmark is diverse and multi-modal, created through collaboration with various hospitals and research institutions.

11.
ArXiv ; 2023 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-37396608

RESUMEN

Gliomas are the most common type of primary brain tumors. Although gliomas are relatively rare, they are among the deadliest types of cancer, with a survival rate of less than 2 years after diagnosis. Gliomas are challenging to diagnose, hard to treat and inherently resistant to conventional therapy. Years of extensive research to improve diagnosis and treatment of gliomas have decreased mortality rates across the Global North, while chances of survival among individuals in low- and middle-income countries (LMICs) remain unchanged and are significantly worse in Sub-Saharan Africa (SSA) populations. Long-term survival with glioma is associated with the identification of appropriate pathological features on brain MRI and confirmation by histopathology. Since 2012, the Brain Tumor Segmentation (BraTS) Challenge have evaluated state-of-the-art machine learning methods to detect, characterize, and classify gliomas. However, it is unclear if the state-of-the-art methods can be widely implemented in SSA given the extensive use of lower-quality MRI technology, which produces poor image contrast and resolution and more importantly, the propensity for late presentation of disease at advanced stages as well as the unique characteristics of gliomas in SSA (i.e., suspected higher rates of gliomatosis cerebri). Thus, the BraTS-Africa Challenge provides a unique opportunity to include brain MRI glioma cases from SSA in global efforts through the BraTS Challenge to develop and evaluate computer-aided-diagnostic (CAD) methods for the detection and characterization of glioma in resource-limited settings, where the potential for CAD tools to transform healthcare are more likely.

12.
Neuroradiology ; 65(9): 1343-1352, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37468750

RESUMEN

PURPOSE: While the T2-FLAIR mismatch sign is highly specific for isocitrate dehydrogenase (IDH)-mutant, 1p/19q-noncodeleted astrocytomas among lower-grade gliomas, its utility in WHO grade 4 gliomas is not well-studied. We derived the partial T2-FLAIR mismatch sign as an imaging biomarker for IDH mutation in WHO grade 4 gliomas. METHODS: Preoperative MRI scans of adult WHO grade 4 glioma patients (n = 2165) from the multi-institutional ReSPOND (Radiomics Signatures for PrecisiON Diagnostics) consortium were analyzed. Diagnostic performance of the partial T2-FLAIR mismatch sign was evaluated. Subset analyses were performed to assess associations of imaging markers with overall survival (OS). RESULTS: One hundred twenty-one (5.6%) of 2165 grade 4 gliomas were IDH-mutant. Partial T2-FLAIR mismatch was present in 40 (1.8%) cases, 32 of which were IDH-mutant, yielding 26.4% sensitivity, 99.6% specificity, 80.0% positive predictive value, and 95.8% negative predictive value. Multivariate logistic regression demonstrated IDH mutation was significantly associated with partial T2-FLAIR mismatch (odds ratio [OR] 5.715, 95% CI [1.896, 17.221], p = 0.002), younger age (OR 0.911 [0.895, 0.927], p < 0.001), tumor centered in frontal lobe (OR 3.842, [2.361, 6.251], p < 0.001), absence of multicentricity (OR 0.173, [0.049, 0.612], p = 0.007), and presence of cystic (OR 6.596, [3.023, 14.391], p < 0.001) or non-enhancing solid components (OR 6.069, [3.371, 10.928], p < 0.001). Multivariate Cox analysis demonstrated cystic components (p = 0.024) and non-enhancing solid components (p = 0.003) were associated with longer OS, while older age (p < 0.001), frontal lobe center (p = 0.008), multifocality (p < 0.001), and multicentricity (p < 0.001) were associated with shorter OS. CONCLUSION: Partial T2-FLAIR mismatch sign is highly specific for IDH mutation in WHO grade 4 gliomas.


Asunto(s)
Neoplasias Encefálicas , Glioma , Adulto , Humanos , Isocitrato Deshidrogenasa/genética , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/genética , Estudios Retrospectivos , Glioma/diagnóstico por imagen , Glioma/genética , Imagen por Resonancia Magnética/métodos , Mutación , Organización Mundial de la Salud
14.
J Digit Imaging ; 36(3): 964-972, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36781588

RESUMEN

Advanced visualization techniques such as maximum intensity projection (MIP) and volume rendering (VR) are useful for evaluating neurovascular anatomy on CT angiography (CTA) of the brain; however, interference from surrounding osseous anatomy is common. Existing methods for removing bone from CTA images are limited in scope and/or accuracy, particularly at the skull base. We present a new brain CTA bone removal tool, which addresses many of these limitations. A deep convolutional neural network was designed and trained for bone removal using 72 brain CTAs. The model was tested on 15 CTAs from the same data source and 17 CTAs from an independent external dataset. Bone removal accuracy was assessed quantitatively, by comparing automated segmentation results to manual segmentations, and qualitatively by evaluating VR visualization of the carotid siphons compared to an existing method for automated bone removal. Average Dice overlap between automated and manual segmentations from the internal and external test datasets were 0.986 and 0.979 respectively. This was superior compared to a publicly available noncontrast head CT bone removal algorithm which had a Dice overlap of 0.947 (internal dataset) and 0.938 (external dataset). Our algorithm yielded better VR visualization of the carotid siphons than the publicly available bone removal tool in 14 out of 15 CTAs (93%, chi-square statistic of 22.5, p-value of < 0.00001) from the internal test dataset and 15 out of 17 CTAs (88%, chi-square statistic of 23.1, p-value of < 0.00001) from the external test dataset. Bone removal allowed subjectively superior MIP and VR visualization of vascular anatomy/pathology. The proposed brain CTA bone removal algorithm is rapid, accurate, and allows superior visualization of vascular anatomy and pathology compared to other available techniques and was validated on an independent external dataset.


Asunto(s)
Angiografía por Tomografía Computarizada , Aprendizaje Profundo , Humanos , Tomografía Computarizada por Rayos X/métodos , Cabeza , Encéfalo/diagnóstico por imagen , Procesamiento de Imagen Asistido por Computador/métodos
16.
Radiology ; 306(3): e213199, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36378030

RESUMEN

Background There is increasing interest in noncontrast breast MRI alternatives for tumor visualization to increase the accessibility of breast MRI. Purpose To evaluate the feasibility and accuracy of generating simulated contrast-enhanced T1-weighted breast MRI scans from precontrast MRI sequences in biopsy-proven invasive breast cancer with use of deep learning. Materials and Methods Women with invasive breast cancer and a contrast-enhanced breast MRI examination that was performed for initial evaluation of the extent of disease between January 2015 and December 2019 at a single academic institution were retrospectively identified. A three-dimensional, fully convolutional deep neural network simulated contrast-enhanced T1-weighted breast MRI scans from five precontrast sequences (T1-weighted non-fat-suppressed [FS], T1-weighted FS, T2-weighted FS, apparent diffusion coefficient, and diffusion-weighted imaging). For qualitative assessment, four breast radiologists (with 3-15 years of experience) blinded to whether the method of contrast was real or simulated assessed image quality (excellent, acceptable, good, poor, or unacceptable), presence of tumor enhancement, and maximum index mass size by using 22 pairs of real and simulated contrast-enhanced MRI scans. Quantitative comparison was performed using whole-breast similarity and error metrics and Dice coefficient analysis of enhancing tumor overlap. Results Ninety-six MRI examinations in 96 women (mean age, 52 years ± 12 [SD]) were evaluated. The readers assessed all simulated MRI scans as having the appearance of a real MRI scan with tumor enhancement. Index mass sizes on real and simulated MRI scans demonstrated good to excellent agreement (intraclass correlation coefficient, 0.73-0.86; P < .001) without significant differences (mean differences, -0.8 to 0.8 mm; P = .36-.80). Almost all simulated MRI scans (84 of 88 [95%]) were considered of diagnostic quality (ratings of excellent, acceptable, or good). Quantitative analysis demonstrated strong similarity (structural similarity index, 0.88 ± 0.05), low voxel-wise error (symmetric mean absolute percent error, 3.26%), and Dice coefficient of enhancing tumor overlap of 0.75 ± 0.25. Conclusion It is feasible to generate simulated contrast-enhanced breast MRI scans with use of deep learning. Simulated and real contrast-enhanced MRI scans demonstrated comparable tumor sizes, areas of tumor enhancement, and image quality without significant qualitative or quantitative differences. © RSNA, 2022 Online supplemental material is available for this article. See also the editorial by Slanetz in this issue. An earlier incorrect version appeared online. This article was corrected on January 17, 2023.


Asunto(s)
Neoplasias de la Mama , Aprendizaje Profundo , Femenino , Humanos , Persona de Mediana Edad , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/patología , Estudios Retrospectivos , Mama/diagnóstico por imagen , Mama/patología , Imagen por Resonancia Magnética/métodos , Medios de Contraste
17.
Radiol Artif Intell ; 4(6): e220058, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36523646

RESUMEN

Supplemental material is available for this article. Keywords: Informatics, MR Diffusion Tensor Imaging, MR Perfusion, MR Imaging, Neuro-Oncology, CNS, Brain/Brain Stem, Oncology, Radiogenomics, Radiology-Pathology Integration © RSNA, 2022.

18.
Nat Commun ; 13(1): 7346, 2022 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-36470898

RESUMEN

Although machine learning (ML) has shown promise across disciplines, out-of-sample generalizability is concerning. This is currently addressed by sharing multi-site data, but such centralization is challenging/infeasible to scale due to various limitations. Federated ML (FL) provides an alternative paradigm for accurate and generalizable ML, by only sharing numerical model updates. Here we present the largest FL study to-date, involving data from 71 sites across 6 continents, to generate an automatic tumor boundary detector for the rare disease of glioblastoma, reporting the largest such dataset in the literature (n = 6, 314). We demonstrate a 33% delineation improvement for the surgically targetable tumor, and 23% for the complete tumor extent, over a publicly trained model. We anticipate our study to: 1) enable more healthcare studies informed by large diverse data, ensuring meaningful results for rare diseases and underrepresented populations, 2) facilitate further analyses for glioblastoma by releasing our consensus model, and 3) demonstrate the FL effectiveness at such scale and task-complexity as a paradigm shift for multi-site collaborations, alleviating the need for data-sharing.


Asunto(s)
Macrodatos , Glioblastoma , Humanos , Aprendizaje Automático , Enfermedades Raras , Difusión de la Información
19.
Radiol Artif Intell ; 4(5): e210243, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36204543

RESUMEN

Neural networks were trained for segmentation and longitudinal assessment of posttreatment diffuse glioma. A retrospective cohort (from January 2018 to December 2019) of 298 patients with diffuse glioma (mean age, 52 years ± 14 [SD]; 177 men; 152 patients with glioblastoma, 72 patients with astrocytoma, and 74 patients with oligodendroglioma) who underwent two consecutive multimodal MRI examinations were randomly selected into training (n = 198) and testing (n = 100) samples. A posttreatment tumor segmentation three-dimensional nnU-Net convolutional neural network with multichannel inputs (T1, T2, and T1 postcontrast and fluid-attenuated inversion recovery [FLAIR]) was trained to segment three multiclass tissue types (peritumoral edematous, infiltrated, or treatment-changed tissue [ED]; active tumor or enhancing tissue [AT]; and necrotic core). Separate longitudinal change nnU-Nets were trained on registered and subtracted FLAIR and T1 postlongitudinal images to localize and better quantify and classify changes in ED and AT. Segmentation Dice scores, volume similarities, and 95th percentile Hausdorff distances ranged from 0.72 to 0.89, 0.90 to 0.96, and 2.5 to 3.6 mm, respectively. Accuracy rates of the posttreatment tumor segmentation and longitudinal change networks being able to classify longitudinal changes in ED and AT as increased, decreased, or unchanged were 76%-79% and 90%-91%, respectively. The accuracy levels of the longitudinal change networks were not significantly different from those of three neuroradiologists (accuracy, 90%-92%; κ, 0.58-0.63; P > .05). The results of this study support the potential clinical value of artificial intelligence-based automated longitudinal assessment of posttreatment diffuse glioma. Keywords: MR Imaging, Neuro-Oncology, Neural Networks, CNS, Brain/Brain Stem, Segmentation, Quantification, Convolutional Neural Network (CNN) Supplemental material is available for this article. © RSNA, 2022.

20.
Neurooncol Adv ; 4(1): vdac060, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35611269

RESUMEN

Background: Glioblastoma is the most common primary brain malignancy, yet treatment options are limited, and prognosis remains guarded. Individualized tumor genetic assessment has become important for accurate prognosis and for guiding emerging targeted therapies. However, challenges remain for widespread tumor genetic testing due to costs and the need for tissue sampling. The aim of this study is to evaluate a novel artificial intelligence method for predicting clinically relevant genetic biomarkers from preoperative brain MRI in patients with glioblastoma. Methods: We retrospectively analyzed preoperative MRI data from 400 patients with glioblastoma, IDH-wildtype or WHO grade 4 astrocytoma, IDH mutant who underwent resection and genetic testing. Nine genetic biomarkers were assessed: hotspot mutations of IDH1 or TERT promoter, pathogenic mutations of TP53, PTEN, ATRX, or CDKN2A/B, MGMT promoter methylation, EGFR amplification, and combined aneuploidy of chromosomes 7 and 10. Models were developed to predict biomarker status from MRI data using radiomics features, convolutional neural network (CNN) features, and a combination of both. Results: Combined model performance was good for IDH1 and TERT promoter hotspot mutations, pathogenic mutations of ATRX and CDKN2A/B, and combined aneuploidy of chromosomes 7 and 10, with receiver operating characteristic area under the curve (ROC AUC) >0.85 and was fair for all other tested biomarkers with ROC AUC >0.7. Combined model performance was statistically superior to individual radiomics and CNN feature models for prediction chromosome 7 and 10 aneuploidy, MGMT promoter methylation, and PTEN mutation. Conclusions: Combining radiomics and CNN features from preoperative MRI yields improved noninvasive genetic biomarker prediction performance in patients with WHO grade 4 diffuse astrocytic gliomas.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...