Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Sci Rep ; 14(1): 4922, 2024 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-38418494

RESUMO

Glioblastoma is a highly heterogeneous disease, with variations observed at both phenotypical and molecular levels. Personalized therapies would be facilitated by non-invasive in vivo approaches for characterizing this heterogeneity. In this study, we developed unsupervised joint machine learning between radiomic and genomic data, thereby identifying distinct glioblastoma subtypes. A retrospective cohort of 571 IDH-wildtype glioblastoma patients were included in the study, and pre-operative multi-parametric MRI scans and targeted next-generation sequencing (NGS) data were collected. L21-norm minimization was used to select a subset of 12 radiomic features from the MRI scans, and 13 key driver genes from the five main signal pathways most affected in glioblastoma were selected from the genomic data. Subtypes were identified using a joint learning approach called Anchor-based Partial Multi-modal Clustering on both radiomic and genomic modalities. Kaplan-Meier analysis identified three distinct glioblastoma subtypes: high-risk, medium-risk, and low-risk, based on overall survival outcome (p < 0.05, log-rank test; Hazard Ratio = 1.64, 95% CI 1.17-2.31, Cox proportional hazard model on high-risk and low-risk subtypes). The three subtypes displayed different phenotypical and molecular characteristics in terms of imaging histogram, co-occurrence of genes, and correlation between the two modalities. Our findings demonstrate the synergistic value of integrated radiomic signatures and molecular characteristics for glioblastoma subtyping. Joint learning on both modalities can aid in better understanding the molecular basis of phenotypical signatures of glioblastoma, and provide insights into the biological underpinnings of tumor formation and progression.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/diagnóstico por imagem , Glioblastoma/genética , Estudos Retrospectivos , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/genética , Prognóstico , Imageamento por Ressonância Magnética/métodos , Genômica
2.
Neurosurg Focus ; 54(6): E17, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37552657

RESUMO

OBJECTIVE: The clinical behavior of meningiomas is not entirely captured by its designated WHO grade, therefore other factors must be elucidated that portend increased tumor aggressiveness and associated risk of recurrence. In this study, the authors identify multiparametric MRI radiomic signatures of meningiomas using Ki-67 as a prognostic marker of clinical outcomes independent of WHO grade. METHODS: A retrospective analysis was conducted of all resected meningiomas between 2012 and 2018. Preoperative MR images were used for high-throughput radiomic feature extraction and subsequently used to develop a machine learning algorithm to stratify meningiomas based on Ki-67 indices < 5% and ≥ 5%, independent of WHO grade. Progression-free survival (PFS) was assessed based on machine learning prediction of Ki-67 strata and compared with outcomes based on histopathological Ki-67. RESULTS: Three hundred forty-three meningiomas were included: 291 with WHO grade I, 43 with grade II, and 9 with grade III. The overall rate of recurrence was 19.8% (15.1% in grade I, 44.2% in grade II, and 77.8% in grade III) over a median follow-up of 28.5 months. Grade II and III tumors had higher Ki-67 indices than grade I tumors, albeit tumor and peritumoral edema volumes had considerable variation independent of meningioma WHO grade. Forty-six high-performing radiomic features (1 morphological, 7 intensity-based, and 38 textural) were identified and used to build a support vector machine model to stratify tumors based on a Ki-67 cutoff of 5%, with resultant areas under the curve of 0.83 (95% CI 0.78-0.89) and 0.84 (95% CI 0.75-0.94) achieved for the discovery (n = 257) and validation (n = 86) data sets, respectively. Comparison of histopathological Ki-67 versus machine learning-predicted Ki-67 showed excellent performance (overall accuracy > 80%), with classification of grade I meningiomas exhibiting the greatest accuracy. Prediction of Ki-67 by machine learning classifier revealed shorter PFS for meningiomas with Ki-67 indices ≥ 5% compared with tumors with Ki-67 < 5% (p < 0.0001, log-rank test), which corroborates divergent patient outcomes observed using histopathological Ki-67. CONCLUSIONS: The Ki-67 proliferation index may serve as a surrogate marker of increased meningioma aggressiveness independent of WHO grade. Machine learning using radiomic feature analysis may be used for the preoperative prediction of meningioma Ki-67, which provides enhanced analytical insights to help improve diagnostic classification and guide patient-specific treatment strategies.


Assuntos
Neoplasias Meníngeas , Meningioma , Humanos , Meningioma/diagnóstico por imagem , Meningioma/cirurgia , Antígeno Ki-67 , Neoplasias Meníngeas/diagnóstico por imagem , Neoplasias Meníngeas/cirurgia , Estudos Retrospectivos , Prognóstico , Proliferação de Células
4.
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
5.
Brainlesion ; 12962: 151-167, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36331281

RESUMO

Brain extraction is an indispensable step in neuro-imaging with a direct impact on downstream analyses. Most such methods have been developed for non-pathologically affected brains, and hence tend to suffer in performance when applied on brains with pathologies, e.g., gliomas, multiple sclerosis, traumatic brain injuries. Deep Learning (DL) methodologies for healthcare have shown promising results, but their clinical translation has been limited, primarily due to these methods suffering from i) high computational cost, and ii) specific hardware requirements, e.g., DL acceleration cards. In this study, we explore the potential of mathematical optimizations, towards making DL methods amenable to application in low resource environments. We focus on both the qualitative and quantitative evaluation of such optimizations on an existing DL brain extraction method, designed for pathologically-affected brains and agnostic to the input modality. We conduct direct optimizations and quantization of the trained model (i.e., prior to inference on new data). Our results yield substantial gains, in terms of speedup, latency, through-put, and reduction in memory usage, while the segmentation performance of the initial and the optimized models remains stable, i.e., as quantified by both the Dice Similarity Coefficient and the Hausdorff Distance. These findings support post-training optimizations as a promising approach for enabling the execution of advanced DL methodologies on plain commercial-grade CPUs, and hence contributing to their translation in limited- and low- resource clinical environments.

6.
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
7.
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
8.
Sci Data ; 9(1): 338, 2022 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-35701399

RESUMO

Malignancy of the brain and CNS is unfortunately a common diagnosis. A large subset of these lesions tends to be high grade tumors which portend poor prognoses and low survival rates, and are estimated to be the tenth leading cause of death worldwide. The complex nature of the brain tissue environment in which these lesions arise offers a rich opportunity for translational research. Magnetic Resonance Imaging (MRI) can provide a comprehensive view of the abnormal regions in the brain, therefore, its applications in the translational brain cancer research is considered essential for the diagnosis and monitoring of disease. Recent years has seen rapid growth in the field of radiogenomics, especially in cancer, and scientists have been able to successfully integrate the quantitative data extracted from medical images (also known as radiomics) with genomics to answer new and clinically relevant questions. In this paper, we took raw MRI scans from the REMBRANDT data collection from public domain, and performed volumetric segmentation to identify subregions of the brain. Radiomic features were then extracted to represent the MRIs in a quantitative yet summarized format. This resulting dataset now enables further biomedical and integrative data analysis, and is being made public via the NeuroImaging Tools & Resources Collaboratory (NITRC) repository ( https://www.nitrc.org/projects/rembrandt_brain/ ).


Assuntos
Neoplasias Encefálicas , Imageamento por Ressonância Magnética , Encéfalo/diagnóstico por imagem , Neoplasias Encefálicas/diagnóstico por imagem , Genômica , Humanos , Neuroimagem
9.
Sci Rep ; 12(1): 8784, 2022 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-35610333

RESUMO

Multi-omic data, i.e., clinical measures, radiomic, and genetic data, capture multi-faceted tumor characteristics, contributing to a comprehensive patient risk assessment. Here, we investigate the additive value and independent reproducibility of integrated diagnostics in prediction of overall survival (OS) in isocitrate dehydrogenase (IDH)-wildtype GBM patients, by combining conventional and deep learning methods. Conventional radiomics and deep learning features were extracted from pre-operative multi-parametric MRI of 516 GBM patients. Support vector machine (SVM) classifiers were trained on the radiomic features in the discovery cohort (n = 404) to categorize patient groups of high-risk (OS < 6 months) vs all, and low-risk (OS ≥ 18 months) vs all. The trained radiomic model was independently tested in the replication cohort (n = 112) and a patient-wise survival prediction index was produced. Multivariate Cox-PH models were generated for the replication cohort, first based on clinical measures solely, and then by layering on radiomics and molecular information. Evaluation of the high-risk and low-risk classifiers in the discovery/replication cohorts revealed area under the ROC curves (AUCs) of 0.78 (95% CI 0.70-0.85)/0.75 (95% CI 0.64-0.79) and 0.75 (95% CI 0.65-0.84)/0.63 (95% CI 0.52-0.71), respectively. Cox-PH modeling showed a concordance index of 0.65 (95% CI 0.6-0.7) for clinical data improving to 0.75 (95% CI 0.72-0.79) for the combination of all omics. This study signifies the value of integrated diagnostics for improved prediction of OS in GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Inteligência Artificial , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Genômica , Glioblastoma/diagnóstico por imagem , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Imageamento por Ressonância Magnética/métodos , Reprodutibilidade dos Testes , Estudos Retrospectivos
10.
Artigo em Inglês | MEDLINE | ID: mdl-36998700

RESUMO

Deep learning (DL) models have provided state-of-the-art performance in various medical imaging benchmarking challenges, including the Brain Tumor Segmentation (BraTS) challenges. However, the task of focal pathology multi-compartment segmentation (e.g., tumor and lesion sub-regions) is particularly challenging, and potential errors hinder translating DL models into clinical workflows. Quantifying the reliability of DL model predictions in the form of uncertainties could enable clinical review of the most uncertain regions, thereby building trust and paving the way toward clinical translation. Several uncertainty estimation methods have recently been introduced for DL medical image segmentation tasks. Developing scores to evaluate and compare the performance of uncertainty measures will assist the end-user in making more informed decisions. In this study, we explore and evaluate a score developed during the BraTS 2019 and BraTS 2020 task on uncertainty quantification (QU-BraTS) and designed to assess and rank uncertainty estimates for brain tumor multi-compartment segmentation. This score (1) rewards uncertainty estimates that produce high confidence in correct assertions and those that assign low confidence levels at incorrect assertions, and (2) penalizes uncertainty measures that lead to a higher percentage of under-confident correct assertions. We further benchmark the segmentation uncertainties generated by 14 independent participating teams of QU-BraTS 2020, all of which also participated in the main BraTS segmentation task. Overall, our findings confirm the importance and complementary value that uncertainty estimates provide to segmentation algorithms, highlighting the need for uncertainty quantification in medical image analyses. Finally, in favor of transparency and reproducibility, our evaluation code is made publicly available at https://github.com/RagMeh11/QU-BraTS.

11.
Neurosurgery ; 89(5): 928-936, 2021 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-34460921

RESUMO

BACKGROUND: Although World Health Organization (WHO) grade I meningiomas are considered "benign" tumors, an elevated Ki-67 is one crucial factor that has been shown to influence tumor behavior and clinical outcomes. The ability to preoperatively discern Ki-67 would confer the ability to guide surgical strategy. OBJECTIVE: In this study, we develop a machine learning (ML) algorithm using radiomic feature analysis to predict Ki-67 in WHO grade I meningiomas. METHODS: A retrospective analysis was performed for a cohort of 306 patients who underwent surgical resection of WHO grade I meningiomas. Preoperative magnetic resonance imaging was used to perform radiomic feature extraction followed by ML modeling using least absolute shrinkage and selection operator wrapped with support vector machine through nested cross-validation on a discovery cohort (n = 230), to stratify tumors based on Ki-67 <5% and ≥5%. The final model was independently tested on a replication cohort (n = 76). RESULTS: An area under the receiver operating curve (AUC) of 0.84 (95% CI: 0.78-0.90) with a sensitivity of 84.1% and specificity of 73.3% was achieved in the discovery cohort. When this model was applied to the replication cohort, a similar high performance was achieved, with an AUC of 0.83 (95% CI: 0.73-0.94), sensitivity and specificity of 82.6% and 85.5%, respectively. The model demonstrated similar efficacy when applied to skull base and nonskull base tumors. CONCLUSION: Our proposed radiomic feature analysis can be used to stratify WHO grade I meningiomas based on Ki-67 with excellent accuracy and can be applied to skull base and nonskull base tumors with similar performance achieved.


Assuntos
Antígeno Ki-67/análise , Neoplasias Meníngeas , Meningioma , Imageamento por Ressonância Magnética Multiparamétrica , Humanos , Aprendizado de Máquina , Imageamento por Ressonância Magnética , Neoplasias Meníngeas/diagnóstico por imagem , Neoplasias Meníngeas/cirurgia , Meningioma/diagnóstico por imagem , Meningioma/cirurgia , Estudos Retrospectivos
12.
Sci Rep ; 11(1): 15011, 2021 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-34294864

RESUMO

Glioblastoma (GBM) has high metabolic demands, which can lead to acidification of the tumor microenvironment. We hypothesize that a machine learning model built on temporal principal component analysis (PCA) of dynamic susceptibility contrast-enhanced (DSC) perfusion MRI can be used to estimate tumor acidity in GBM, as estimated by pH-sensitive amine chemical exchange saturation transfer echo-planar imaging (CEST-EPI). We analyzed 78 MRI scans in 32 treatment naïve and post-treatment GBM patients. All patients were imaged with DSC-MRI, and pH-weighting that was quantified from CEST-EPI estimation of the magnetization transfer ratio asymmetry (MTRasym) at 3 ppm. Enhancing tumor (ET), non-enhancing core (NC), and peritumoral T2 hyperintensity (namely, edema, ED) were used to extract principal components (PCs) and to build support vector machines regression (SVR) models to predict MTRasym values using PCs. Our predicted map correlated with MTRasym values with Spearman's r equal to 0.66, 0.47, 0.67, 0.71, in NC, ET, ED, and overall, respectively (p < 0.006). The results of this study demonstrates that PCA analysis of DSC imaging data can provide information about tumor pH in GBM patients, with the strongest association within the peritumoral regions.


Assuntos
Glioblastoma/diagnóstico por imagem , Glioblastoma/patologia , Concentração de Íons de Hidrogênio , Imageamento por Ressonância Magnética/métodos , Microambiente Tumoral , Idoso , Animais , Interpretação Estatística de Dados , Modelos Animais de Doenças , Feminino , Humanos , Processamento de Imagem Assistida por Computador , Imageamento por Ressonância Magnética/normas , Masculino , Camundongos , Pessoa de Meia-Idade , Gradação de Tumores , Estadiamento de Neoplasias , Análise de Componente Principal
13.
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
14.
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
15.
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
18.
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.

19.
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.

20.
Brainlesion ; 11992: 57-68, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32577629

RESUMO

Skull-stripping is an essential pre-processing step in computational neuro-imaging directly impacting subsequent analyses. Existing skull-stripping methods have primarily targeted non-pathologicallyaffected brains. Accordingly, they may perform suboptimally when applied on brain Magnetic Resonance Imaging (MRI) scans that have clearly discernible 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. Here we present a performance evaluation of publicly available implementations of established 3D Deep Learning architectures for semantic segmentation (namely DeepMedic, 3D U-Net, FCN), with a particular focus on identifying a skull-stripping approach that performs well on brain tumor scans, and also has a low computational footprint. We have identified a retrospective dataset of 1,796 mpMRI brain tumor scans, with corresponding manually-inspected and verified gold-standard brain tissue segmentations, acquired during standard clinical practice under varying acquisition protocols at the Hospital of the University of Pennsylvania. Our quantitative evaluation identified DeepMedic as the best performing method (Dice = 97.9, Hausdorf f 95 = 2.68). We release this pre-trained model through the Cancer Imaging Phenomics Toolkit (CaPTk) platform.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA