Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
ArXiv ; 2023 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-37608932

RESUMO

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.

3.
Nat Commun ; 13(1): 7346, 2022 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-36470898

RESUMO

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.


Assuntos
Big Data , Glioblastoma , Humanos , Aprendizado de Máquina , Doenças Raras , Disseminação de Informação
4.
Phys Med Biol ; 67(20)2022 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-36137534

RESUMO

Objective.De-centralized data analysis becomes an increasingly preferred option in the healthcare domain, as it alleviates the need for sharing primary patient data across collaborating institutions. This highlights the need for consistent harmonized data curation, pre-processing, and identification of regions of interest based on uniform criteria.Approach.Towards this end, this manuscript describes theFederatedTumorSegmentation (FeTS) tool, in terms of software architecture and functionality.Main results.The primary aim of the FeTS tool is to facilitate this harmonized processing and the generation of gold standard reference labels for tumor sub-compartments on brain magnetic resonance imaging, and further enable federated training of a tumor sub-compartment delineation model across numerous sites distributed across the globe, without the need to share patient data.Significance.Building upon existing open-source tools such as the Insight Toolkit and Qt, the FeTS tool is designed to enable training deep learning models targeting tumor delineation in either centralized or federated settings. The target audience of the FeTS tool is primarily the computational researcher interested in developing federated learning models, and interested in joining a global federation towards this effort. The tool is open sourced athttps://github.com/FETS-AI/Front-End.


Assuntos
Neoplasias , Software , Encéfalo , Humanos , Imageamento por Ressonância Magnética/métodos
5.
Sci Data ; 9(1): 453, 2022 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-35906241

RESUMO

Glioblastoma is the most common aggressive adult brain tumor. Numerous studies have reported results from either private institutional data or publicly available datasets. However, current public datasets are limited in terms of: a) number of subjects, b) lack of consistent acquisition protocol, c) data quality, or d) accompanying clinical, demographic, and molecular information. Toward alleviating these limitations, we contribute the "University of Pennsylvania Glioblastoma Imaging, Genomics, and Radiomics" (UPenn-GBM) dataset, which describes the currently largest publicly available comprehensive collection of 630 patients diagnosed with de novo glioblastoma. The UPenn-GBM dataset includes (a) advanced multi-parametric magnetic resonance imaging scans acquired during routine clinical practice, at the University of Pennsylvania Health System, (b) accompanying clinical, demographic, and molecular information, (d) perfusion and diffusion derivative volumes, (e) computationally-derived and manually-revised expert annotations of tumor sub-regions, as well as (f) quantitative imaging (also known as radiomic) features corresponding to each of these regions. This collection describes our contribution towards repeatable, reproducible, and comparative quantitative studies leading to new predictive, prognostic, and diagnostic assessments.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Adulto , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/fisiopatologia , Genômica , Glioblastoma/diagnóstico por imagem , Glioblastoma/genética , Glioblastoma/fisiopatologia , Humanos , Imageamento por Ressonância Magnética , Prognóstico
6.
Nat Commun ; 13(1): 4128, 2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35840566

RESUMO

International challenges have become the de facto standard for comparative assessment of image analysis algorithms. Although segmentation is the most widely investigated medical image processing task, the various challenges have been organized to focus only on specific clinical tasks. We organized the Medical Segmentation Decathlon (MSD)-a biomedical image analysis challenge, in which algorithms compete in a multitude of both tasks and modalities to investigate the hypothesis that a method capable of performing well on multiple tasks will generalize well to a previously unseen task and potentially outperform a custom-designed solution. MSD results confirmed this hypothesis, moreover, MSD winner continued generalizing well to a wide range of other clinical problems for the next two years. Three main conclusions can be drawn from this study: (1) state-of-the-art image segmentation algorithms generalize well when retrained on unseen tasks; (2) consistent algorithmic performance across multiple tasks is a strong surrogate of algorithmic generalizability; (3) the training of accurate AI segmentation models is now commoditized to scientists that are not versed in AI model training.


Assuntos
Algoritmos , Processamento de Imagem Assistida por Computador , Processamento de Imagem Assistida por Computador/métodos
7.
Front Med (Lausanne) ; 9: 797586, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35372431

RESUMO

Multiple Sclerosis (MS) is a demyelinating disease of the central nervous system that affects nearly 1 million adults in the United States. Magnetic Resonance Imaging (MRI) plays a vital role in diagnosis and treatment monitoring in MS patients. In particular, follow-up MRI with T2-FLAIR images of the brain, depicting white matter lesions, is the mainstay for monitoring disease activity and making treatment decisions. In this article, we present a computational approach that has been deployed and integrated into a real-world routine clinical workflow, focusing on two tasks: (a) detecting new disease activity in MS patients, and (b) determining the necessity for injecting Gadolinium Based Contract Agents (GBCAs). This computer-aided detection (CAD) software has been utilized for the former task on more than 19, 000 patients over the course of 10 years, while its added function of identifying patients who need GBCA injection, has been operative for the past 3 years, with > 85% sensitivity. The benefits of this approach are summarized in: (1) offering a reproducible and accurate clinical assessment of MS lesion patients, (2) reducing the adverse effects of GBCAs (and the deposition of GBCAs to the patient's brain) by identifying the patients who may benefit from injection, and (3) reducing healthcare costs, patients' discomfort, and caregivers' workload.

8.
J Neuroimaging ; 32(4): 656-666, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35294074

RESUMO

BACKGROUND AND PURPOSE: Imaging and autopsy studies show intracranial gadolinium deposition in patients who have undergone serial contrast-enhanced MRIs. This observation has raised concerns when using contrast administration in patients who receive frequent MRIs. To address this, we implemented a contrast-conditional protocol wherein gadolinium is administered only for multiple sclerosis (MS) patients with imaging evidence of new disease activity on precontrast imaging. In this study, we explore the economic impact of our new MRI protocol. METHODS: We compared scanner time and Medicare reimbursement using our contrast-conditional methodology versus that of prior protocols where all patients received gadolinium. RESULTS: For 422 patients over 4 months, the contrast-conditional protocol amounted to 60% decrease in contrast injection and savings of approximately 20% of MRI scanner time. If the extra scanner time was used for performing MS follow-up MRIs in additional patients, the contrast-conditional protocol would amount to net revenue loss of $21,707 (∼3.7%). CONCLUSIONS: Implementation of a new protocol to limit contrast in MS follow-up MRIs led to a minimal decrease in revenue when controlled for scanner time utilized and is outweighed by other benefits, including substantial decreased gadolinium administration, increased patient comfort, and increased availability of scanner time, which depending on type of studies performed could result in additional financial benefit.


Assuntos
Gadolínio , Esclerose Múltipla , Idoso , Meios de Contraste , Seguimentos , Humanos , Imageamento por Ressonância Magnética/métodos , Medicare , Esclerose Múltipla/diagnóstico por imagem , Estados Unidos
9.
Appl Sci (Basel) ; 11(16)2021 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-34621541

RESUMO

We seek the development and evaluation of a fast, accurate, and consistent method for general-purpose segmentation, based on interactive machine learning (IML). To validate our method, we identified retrospective cohorts of 20 brain, 50 breast, and 50 lung cancer patients, as well as 20 spleen scans, with corresponding ground truth annotations. Utilizing very brief user training annotations and the adaptive geodesic distance transform, an ensemble of SVMs is trained, providing a patient-specific model applied to the whole image. Two experts segmented each cohort twice with our method and twice manually. The IML method was faster than manual annotation by 53.1% on average. We found significant (p < 0.001) overlap difference for spleen (DiceIML/DiceManual = 0.91/0.87), breast tumors (DiceIML/DiceManual = 0.84/0.82), and lung nodules (DiceIML/DiceManual = 0.78/0.83). For intra-rater consistency, a significant (p = 0.003) difference was found for spleen (DiceIML/DiceManual = 0.91/0.89). For inter-rater consistency, significant (p < 0.045) differences were found for spleen (DiceIML/DiceManual = 0.91/0.87), breast (DiceIML/DiceManual = 0.86/0.81), lung (DiceIML/DiceManual = 0.85/0.89), the non-enhancing (DiceIML/DiceManual = 0.79/0.67) and the enhancing (DiceIML/DiceManual = 0.79/0.84) brain tumor sub-regions, which, in aggregation, favored our method. Quantitative evaluation for speed, spatial overlap, and consistency, reveals the benefits of our proposed method when compared with manual annotation, for several clinically relevant problems. We publicly release our implementation through CaPTk (Cancer Imaging Phenomics Toolkit) and as an MITK plugin.

10.
Stroke ; 52(11): 3419-3426, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34455822

RESUMO

Background and Purpose: Randomized patent foramen ovale closure trials have used open-label end point ascertainment which increases the risk of bias and undermines confidence in the conclusions. The Gore REDUCE trial prospectively performed baseline and follow-up magnetic resonance imaging (MRIs) for all subjects providing an objective measure of the effectiveness of closure. Methods: We performed blinded evaluations of the presence, location, and volume of new infarct on diffusion-weighted imaging of recurrent clinical stroke or new infarct (>3 mm) on T2/fluid attenuated inversion recovery from baseline to follow-up MRI at 2 years, comparing closure to medical therapy alone. We also examined the effect of shunt size and the development of atrial fibrillation on infarct burden at follow-up. Results: At follow-up, new clinical stroke or silent MRI infarct occurred in 18/383 (4.7%) patients who underwent closure and 19/177 (10.7%) medication-only patients (relative risk, 0.44 [95% CI, 0.24­0.81], P=0.02). Clinical strokes were less common in closure patients compared with medically treated patients, 5 (1.3%) versus 12 (6.8%), P=0.001, while silent MRI infarcts were similar, 13 (3.4%) versus 7 (4.0%), P=0.81. There were no differences in number, volumes, and distribution of new infarct comparing closure patients to those treated with medication alone. There were also no differences of number, volumes, and distribution comparing silent infarcts to clinical strokes. Infarct burden was also similar for patients who developed atrial fibrillation and for those with large shunts. Conclusions: The REDUCE trial demonstrates that patent foramen ovale closure prevents recurrent brain infarction based on the objective outcome of new infarcts on MRI. Only clinical strokes were reduced by closure while silent infarctions were similar between study arms, and there were no differences in infarct volume or location comparing silent infarcts to clinical strokes. Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT00738894.


Assuntos
Infarto Encefálico/epidemiologia , Infarto Encefálico/patologia , Forame Oval Patente/complicações , Forame Oval Patente/cirurgia , Infarto Encefálico/etiologia , Humanos , Incidência , Imageamento por Ressonância Magnética
11.
Radiol Imaging Cancer ; 3(4): e200108, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34296969

RESUMO

Purpose To test the hypothesis that combined features from MR and digital histopathologic images more accurately predict overall survival (OS) in patients with glioma compared with MRI or histopathologic features alone. Materials and Methods Multiparametric MR and histopathologic images in patients with a diagnosis of glioma (high- or low-grade glioma [HGG or LGG]) were obtained from The Cancer Imaging Archive (original images acquired 1983-2008). An extensive set of engineered features such as intensity, histogram, and texture were extracted from delineated tumor regions in MR and histopathologic images. Cox proportional hazard regression and support vector machine classification (SVC) models were applied to (a) MRI features only (MRIcox/svc), histopathologic features only (HistoPathcox/svc), and (c) combined MRI and histopathologic features (MRI+HistoPathcox/svc) and evaluated in a split train-test configuration. Results A total of 171 patients (mean age, 51 years ± 15; 91 men) were included with HGG (n = 75) and LGG (n = 96). Median OS was 467 days (range, 3-4752 days) for all patients, 350 days (range, 15-1561 days) for HGG, and 595 days (range, 3-4752 days) for LGG. The MRI+HistoPathcox model demonstrated higher concordance index (C-index) compared with MRIcox and HistoPathcox models on all patients (C-index, 0.79 vs 0.70 [P = .02; MRIcox] and 0.67 [P = .01; HistoPathcox]), patients with HGG (C-index, 0.78 vs 0.68 [P = .03; MRIcox] and 0.64 [P = .01; HistoPathcox]), and patients with LGG (C-index, 0.88 vs 0.62 [P = .008; MRIcox] and 0.62 [P = .006; HistoPathcox]). In binary classification, the MRI+HistoPathsvc model (area under the receiver operating characteristic curve [AUC], 0.86 [95% CI: 0.80, 0.95]) had higher performance than the MRIsvc model (AUC, 0.68 [95% CI: 0.50, 0.81]; P = .01) and the HistoPathsvc model (AUC, 0.72 [95% CI: 0.60, 0.85]; P = .04). Conclusion The model combining features from MR and histopathologic images had higher accuracy in predicting OS compared with the models with MR or histopathologic images alone. Keywords: Survival Prediction, Gliomas, Digital Pathology Imaging, MR Imaging, Machine Learning Supplemental material is available for this article.


Assuntos
Neoplasias Encefálicas , Glioma , Neoplasias Encefálicas/diagnóstico por imagem , Glioma/diagnóstico por imagem , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Máquina de Vetores de Suporte
12.
Med Phys ; 47(12): 6039-6052, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33118182

RESUMO

PURPOSE: The availability of radiographic magnetic resonance imaging (MRI) scans for the Ivy Glioblastoma Atlas Project (Ivy GAP) has opened up opportunities for development of radiomic markers for prognostic/predictive applications in glioblastoma (GBM). In this work, we address two critical challenges with regard to developing robust radiomic approaches: (a) the lack of availability of reliable segmentation labels for glioblastoma tumor sub-compartments (i.e., enhancing tumor, non-enhancing tumor core, peritumoral edematous/infiltrated tissue) and (b) identifying "reproducible" radiomic features that are robust to segmentation variability across readers/sites. ACQUISITION AND VALIDATION METHODS: From TCIA's Ivy GAP cohort, we obtained a paired set (n = 31) of expert annotations approved by two board-certified neuroradiologists at the Hospital of the University of Pennsylvania (UPenn) and at Case Western Reserve University (CWRU). For these studies, we performed a reproducibility study that assessed the variability in (a) segmentation labels and (b) radiomic features, between these paired annotations. The radiomic variability was assessed on a comprehensive panel of 11 700 radiomic features including intensity, volumetric, morphologic, histogram-based, and textural parameters, extracted for each of the paired sets of annotations. Our results demonstrated (a) a high level of inter-rater agreement (median value of DICE ≥0.8 for all sub-compartments), and (b) ≈24% of the extracted radiomic features being highly correlated (based on Spearman's rank correlation coefficient) to annotation variations. These robust features largely belonged to morphology (describing shape characteristics), intensity (capturing intensity profile statistics), and COLLAGE (capturing heterogeneity in gradient orientations) feature families. DATA FORMAT AND USAGE NOTES: We make publicly available on TCIA's Analysis Results Directory (https://doi.org/10.7937/9j41-7d44), the complete set of (a) multi-institutional expert annotations for the tumor sub-compartments, (b) 11 700 radiomic features, and (c) the associated reproducibility meta-analysis. POTENTIAL APPLICATIONS: The annotations and the associated meta-data for Ivy GAP are released with the purpose of enabling researchers toward developing image-based biomarkers for prognostic/predictive applications in GBM.


Assuntos
Glioblastoma , Estudos de Coortes , Glioblastoma/diagnóstico por imagem , Humanos , Imageamento por Ressonância Magnética , Reprodutibilidade dos Testes
13.
Neuroimage ; 220: 117081, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32603860

RESUMO

Brain extraction, or skull-stripping, is an essential pre-processing step in neuro-imaging that has a direct impact on the quality of all subsequent processing and analyses steps. It is also a key requirement in multi-institutional collaborations to comply with privacy-preserving regulations. Existing automated methods, including Deep Learning (DL) based methods that have obtained state-of-the-art results in recent years, have primarily targeted brain extraction without considering pathologically-affected brains. Accordingly, they perform sub-optimally when applied on magnetic resonance imaging (MRI) brain scans with apparent pathologies such as brain tumors. Furthermore, existing methods focus on using only T1-weighted MRI scans, even though multi-parametric MRI (mpMRI) scans are routinely acquired for patients with suspected brain tumors. In this study, we present a comprehensive performance evaluation of recent deep learning architectures for brain extraction, training models on mpMRI scans of pathologically-affected brains, with a particular focus on seeking a practically-applicable, low computational footprint approach, generalizable across multiple institutions, further facilitating collaborations. We identified a large retrospective multi-institutional dataset of n=3340 mpMRI brain tumor scans, with manually-inspected and approved gold-standard segmentations, acquired during standard clinical practice under varying acquisition protocols, both from private institutional data and public (TCIA) collections. To facilitate optimal utilization of rich mpMRI data, we further introduce and evaluate a novel ''modality-agnostic training'' technique that can be applied using any available modality, without need for model retraining. Our results indicate that the modality-agnostic approach1 obtains accurate results, providing a generic and practical tool for brain extraction on scans with brain tumors.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Encéfalo/diagnóstico por imagem , Glioma/diagnóstico por imagem , Processamento de Imagem Assistida por Computador/métodos , Imageamento por Ressonância Magnética/métodos , Bases de Dados Factuais , Aprendizado Profundo , Humanos , Estudos Retrospectivos
14.
J Med Imaging (Bellingham) ; 7(3): 031505, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32566694

RESUMO

Purpose: Glioblastoma, the most common and aggressive adult brain tumor, is considered noncurative at diagnosis, with 14 to 16 months median survival following treatment. There is increasing evidence that noninvasive integrative analysis of radiomic features can predict overall and progression-free survival, using advanced multiparametric magnetic resonance imaging (Adv-mpMRI). If successfully applicable, such noninvasive markers can considerably influence patient management. However, most patients prior to initiation of therapy typically undergo only basic structural mpMRI (Bas-mpMRI, i.e., T1, T1-Gd, T2, and T2-fluid-attenuated inversion recovery) preoperatively, rather than Adv-mpMRI that provides additional vascularization (dynamic susceptibility contrast-MRI) and cell-density (diffusion tensor imaging) related information. Approach: We assess a retrospective cohort of 101 glioblastoma patients with available Adv-mpMRI from a previous study, which has shown that an initial feature panel (IFP, i.e., intensity, volume, location, and growth model parameters) extracted from Adv-mpMRI can yield accurate overall survival stratification. We focus on demonstrating that equally accurate prediction models can be constructed using augmented radiomic feature panels (ARFPs, i.e., integrating morphology and textural descriptors) extracted solely from widely available Bas-mpMRI, obviating the need for using Adv-mpMRI. We extracted 1612 radiomic features from distinct tumor subregions to build multivariate models that stratified patients as long-, intermediate-, or short-survivors. Results: The classification accuracy of the model utilizing Adv-mpMRI protocols and the IFP was 72.77% and degraded to 60.89% when using only Bas-mpMRI. However, utilizing the ARFP on Bas-mpMRI improved the accuracy to 74.26%. Furthermore, Kaplan-Meier analysis demonstrated superior classification of subjects into short-, intermediate-, and long-survivor classes when using ARFP extracted from Bas-mpMRI. Conclusions: This quantitative evaluation indicates that accurate survival prediction in glioblastoma patients is feasible using solely Bas-mpMRI and integrative advanced radiomic features, which can compensate for the lack of Adv-mpMRI. Our finding holds promise for generalization across multiple institutions that may not have access to Adv-mpMRI and to better inform clinical decision-making about aggressive interventions and clinical trials.

16.
JCO Clin Cancer Inform ; 4: 234-244, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32191542

RESUMO

PURPOSE: To construct a multi-institutional radiomic model that supports upfront prediction of progression-free survival (PFS) and recurrence pattern (RP) in patients diagnosed with glioblastoma multiforme (GBM) at the time of initial diagnosis. PATIENTS AND METHODS: We retrospectively identified data for patients with newly diagnosed GBM from two institutions (institution 1, n = 65; institution 2, n = 15) who underwent gross total resection followed by standard adjuvant chemoradiation therapy, with pathologically confirmed recurrence, sufficient follow-up magnetic resonance imaging (MRI) scans to reliably determine PFS, and available presurgical multiparametric MRI (MP-MRI). The advanced software suite Cancer Imaging Phenomics Toolkit (CaPTk) was leveraged to analyze standard clinical brain MP-MRI scans. A rich set of imaging features was extracted from the MP-MRI scans acquired before the initial resection and was integrated into two distinct imaging signatures for predicting mean shorter or longer PFS and near or distant RP. The predictive signatures for PFS and RP were evaluated on the basis of different classification schemes: single-institutional analysis, multi-institutional analysis with random partitioning of the data into discovery and replication cohorts, and multi-institutional assessment with data from institution 1 as the discovery cohort and data from institution 2 as the replication cohort. RESULTS: These predictors achieved cross-validated classification performance (ie, area under the receiver operating characteristic curve) of 0.88 (single-institution analysis) and 0.82 to 0.83 (multi-institution analysis) for prediction of PFS and 0.88 (single-institution analysis) and 0.56 to 0.71 (multi-institution analysis) for prediction of RP. CONCLUSION: Imaging signatures of presurgical MP-MRI scans reveal relatively high predictability of time and location of GBM recurrence, subject to the patients receiving standard first-line chemoradiation therapy. Through its graphical user interface, CaPTk offers easy accessibility to advanced computational algorithms for deriving imaging signatures predictive of clinical outcome and could similarly be used for a variety of radiomic and radiogenomic analyses.


Assuntos
Neoplasias Encefálicas/mortalidade , Glioblastoma/mortalidade , Interpretação de Imagem Assistida por Computador/métodos , Imageamento por Ressonância Magnética Multiparamétrica/métodos , Recidiva Local de Neoplasia/mortalidade , Fenômica/métodos , Software , Adulto , Idoso , Idoso de 80 Anos ou mais , Algoritmos , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/cirurgia , Feminino , Glioblastoma/metabolismo , Glioblastoma/patologia , Glioblastoma/cirurgia , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/cirurgia , Intervalo Livre de Progressão , Curva ROC , Estudos Retrospectivos , Taxa de Sobrevida , Adulto Jovem
17.
Cancer ; 126(11): 2625-2636, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32129893

RESUMO

BACKGROUND: Imaging of glioblastoma patients after maximal safe resection and chemoradiation commonly demonstrates new enhancements that raise concerns about tumor progression. However, in 30% to 50% of patients, these enhancements primarily represent the effects of treatment, or pseudo-progression (PsP). We hypothesize that quantitative machine learning analysis of clinically acquired multiparametric magnetic resonance imaging (mpMRI) can identify subvisual imaging characteristics to provide robust, noninvasive imaging signatures that can distinguish true progression (TP) from PsP. METHODS: We evaluated independent discovery (n = 40) and replication (n = 23) cohorts of glioblastoma patients who underwent second resection due to progressive radiographic changes suspicious for recurrence. Deep learning and conventional feature extraction methods were used to extract quantitative characteristics from the mpMRI scans. Multivariate analysis of these features revealed radiophenotypic signatures distinguishing among TP, PsP, and mixed response that compared with similar categories blindly defined by board-certified neuropathologists. Additionally, interinstitutional validation was performed on 20 new patients. RESULTS: Patients who demonstrate TP on neuropathology are significantly different (P < .0001) from those with PsP, showing imaging features reflecting higher angiogenesis, higher cellularity, and lower water concentration. The accuracy of the proposed signature in leave-one-out cross-validation was 87% for predicting PsP (area under the curve [AUC], 0.92) and 84% for predicting TP (AUC, 0.83), whereas in the discovery/replication cohort, the accuracy was 87% for predicting PsP (AUC, 0.84) and 78% for TP (AUC, 0.80). The accuracy in the interinstitutional cohort was 75% (AUC, 0.80). CONCLUSION: Quantitative mpMRI analysis via machine learning reveals distinctive noninvasive signatures of TP versus PsP after treatment of glioblastoma. Integration of the proposed method into clinical studies can be performed using the freely available Cancer Imaging Phenomics Toolkit.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Aprendizado de Máquina , Imageamento por Ressonância Magnética/métodos , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais , Neoplasias Encefálicas/diagnóstico por imagem , Progressão da Doença , Feminino , Glioblastoma/diagnóstico por imagem , Humanos , Masculino , Pessoa de Meia-Idade
18.
Artigo em Inglês | MEDLINE | ID: mdl-33746333

RESUMO

Glioblastoma, the most common and aggressive adult brain tumor, is considered non-curative at diagnosis. Current literature shows promise on imaging-based overall survival prediction for patients with glioblastoma while integrating advanced (structural, perfusion, and diffusion) multipara metric magnetic resonance imaging (Adv-mpMRI). However, most patients prior to initiation of therapy typically undergo only basic structural mpMRI (Bas-mpMRI, i.e., T1,T1-Gd,T2,T2-FLAIR) pre-operatively, rather than Adv-mpMRI. Here we assess a retrospective cohort of 101 glioblastoma patients with available Adv-mpMRI from a previous study, which has shown that an initial feature panel (IFP) extracted from Adv-mpMRI can yield accurate overall survival stratification. We further focus on demonstrating that equally accurate prediction models can be constructed using augmented feature panels (AFP) extracted solely from Bas-mpMRI, obviating the need for using Adv-mpMRI. The classification accuracy of the model utilizing Adv-mpMRI protocols and the IFP was 72.77%, and improved to 74.26% when utilizing the AFP on Bas-mpMRI. Furthermore, Kaplan-Meier analysis demonstrated superior classification of subjects into short-, intermediate-, and long-survivor classes when using AFPon Basic-mpMRI. This quantitative evaluation indicates that accurate survival prediction in glioblastoma patients is feasible by using solely Bas-mpMRI and integrative radiomic analysis can compensate for the lack of Adv-mpMRI. Our finding holds promise for predicting overall survival based on commonly-acquired Bas-mpMRI, and hence for potential generalization across multiple institutions that may not have access to Adv-mpMRI, facilitating better patient selection.

19.
Neurooncol Adv ; 2(Suppl 4): iv22-iv34, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33521638

RESUMO

BACKGROUND: Gliomas represent a biologically heterogeneous group of primary brain tumors with uncontrolled cellular proliferation and diffuse infiltration that renders them almost incurable, thereby leading to a grim prognosis. Recent comprehensive genomic profiling has greatly elucidated the molecular hallmarks of gliomas, including the mutations in isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2), loss of chromosomes 1p and 19q (1p/19q), and epidermal growth factor receptor variant III (EGFRvIII). Detection of these molecular alterations is based on ex vivo analysis of surgically resected tissue specimen that sometimes is not adequate for testing and/or does not capture the spatial tumor heterogeneity of the neoplasm. METHODS: We developed a method for noninvasive detection of radiogenomic markers of IDH both in lower-grade gliomas (WHO grade II and III tumors) and glioblastoma (WHO grade IV), 1p/19q in IDH-mutant lower-grade gliomas, and EGFRvIII in glioblastoma. Preoperative MRIs of 473 glioma patients from 3 of the studies participating in the ReSPOND consortium (collection I: Hospital of the University of Pennsylvania [HUP: n = 248], collection II: The Cancer Imaging Archive [TCIA; n = 192], and collection III: Ohio Brain Tumor Study [OBTS, n = 33]) were collected. Neuro-Cancer Imaging Phenomics Toolkit (neuro-CaPTk), a modular platform available for cancer imaging analytics and machine learning, was leveraged to extract histogram, shape, anatomical, and texture features from delineated tumor subregions and to integrate these features using support vector machine to generate models predictive of IDH, 1p/19q, and EGFRvIII. The models were validated using 3 configurations: (1) 70-30% training-testing splits or 10-fold cross-validation within individual collections, (2) 70-30% training-testing splits within merged collections, and (3) training on one collection and testing on another. RESULTS: These models achieved a classification accuracy of 86.74% (HUP), 85.45% (TCIA), and 75.15% (TCIA) in identifying EGFRvIII, IDH, and 1p/19q, respectively, in configuration I. The model, when applied on combined data in configuration II, yielded a classification success rate of 82.50% in predicting IDH mutation (HUP + TCIA + OBTS). The model when trained on TCIA dataset yielded classification accuracy of 84.88% in predicting IDH in HUP dataset. CONCLUSIONS: Using machine learning algorithms, high accuracy was achieved in the prediction of IDH, 1p/19q, and EGFRvIII mutation. Neuro-CaPTk encompasses all the pipelines required to replicate these analyses in multi-institutional settings and could also be used for other radio(geno)mic analyses.

20.
Mach Learn Med Imaging ; 12436: 342-352, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34382033

RESUMO

Registration of images with pathologies is challenging due to tissue appearance changes and missing correspondences caused by the pathologies. Moreover, mass effects as observed for brain tumors may displace tissue, creating larger deformations over time than what is observed in a healthy brain. Deep learning models have successfully been applied to image registration to offer dramatic speed up and to use surrogate information (e.g., segmentations) during training. However, existing approaches focus on learning registration models using images from healthy patients. They are therefore not designed for the registration of images with strong pathologies for example in the context of brain tumors, and traumatic brain injuries. In this work, we explore a deep learning approach to register images with brain tumors to an atlas. Our model learns an appearance mapping from images with tumors to the atlas, while simultaneously predicting the transformation to atlas space. Using separate decoders, the network disentangles the tumor mass effect from the reconstruction of quasi-normal images. Results on both synthetic and real brain tumor scans show that our approach outperforms cost function masking for registration to the atlas and that reconstructed quasi-normal images can be used for better longitudinal registrations.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...