Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-38604733

RESUMEN

BACKGROUND AND PURPOSE: Feature variability in radiomics studies due to technical and magnet strength parameters is well known and may be addressed through various pre-processing methods. However, very few studies have evaluated downstream impact of variable pre-processing on model classification performance in a multi-class setting. We sought to evaluate the impact of SUSAN denoising and ComBat harmonization on model classification performance. MATERIALS AND METHODS: A total of 493 cases (410 internal and 83 external dataset) of glioblastoma (GB), intracranial metastatic disease (IMD) and primary CNS lymphoma (PCNSL) underwent semi-automated 3D-segmentation post baseline image processing (BIP) consisting of resampling, realignment, co-registration, skull stripping and image normalization. Post BIP, two sets were generated, one with and another without SUSAN denoising (SD). Radiomics features were extracted from both datasets and batch corrected to produce four datasets: (a) BIP, (b) BIP with SD, (c) BIP with ComBat and (d) BIP with both SD and ComBat harmonization. Performance was then summarized for models using a combination of six feature selection techniques and six machine learning models across four mask-sequence combinations with features derived from one-three (multi-parametric) MRI sequences. RESULTS: Most top performing models on the external test set used BIP+SD derived features. Overall, use of SD and ComBat harmonization led to a slight but generally consistent improvement in model performance on the external test set. CONCLUSIONS: The use of image pre-processing steps such as SD and ComBat harmonization may be more useful in a multiinstitutional setting and improve model generalizability. Models derived from only T1-CE images showed comparable performance to models derived from multiparametric MRI.

2.
Acad Radiol ; 2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-37977889

RESUMEN

RATIONALE AND OBJECTIVES: Imaging-based differentiation between glioblastoma (GB) and brain metastases (BM) remains challenging. Our aim was to evaluate the performance of 3D-convolutional neural networks (CNN) to address this binary classification problem. MATERIALS AND METHODS: T1-CE, T2WI, and FLAIR 3D-segmented masks of 307 patients (157 GB and 150 BM) were generated post resampling, co-registration normalization and semi-automated 3D-segmentation and used for internal model development. Subsequent external validation was performed on 59 cases (27 GB and 32 BM) from another institution. Four different mask-sequence combinations were evaluated using area under the curve (AUC), precision, recall and F1-scores. Diagnostic performance of a neuroradiologist and a general radiologist, both without and with the model output available, was also assessed. RESULTS: 3D-model using the T1-CE tumor mask (TM) showed the highest performance [AUC 0.93 (95% CI 0.858-0.995)] on the external test set, followed closely by the model using T1-CE TM and FLAIR mask of peri-tumoral region (PTR) [AUC of 0.91 (95% CI 0.834-0.986)]. Models using T2WI masks showed robust performance on the internal dataset but lower performance on the external set. Both neuroradiologist and general radiologist showed improved performance with model output provided [AUC increased from 0.89 to 0.968 (p = 0.06) and from 0.78 to 0.965 (p = 0.007) respectively], the latter being statistically significant. CONCLUSION: 3D-CNNs showed robust performance for differentiating GB from BMs, with T1-CE TM, either alone or combined with FLAIR-PTR masks. Availability of model output significantly improved the accuracy of the general radiologist.

3.
JAMA Neurol ; 80(12): 1295-1306, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37930695

RESUMEN

Importance: For the Alzheimer disease (AD) therapies to effectively attenuate clinical progression, it may be critical to intervene before the onset of amyloid-associated tau spreading, which drives neurodegeneration and cognitive decline. Time points at which amyloid-associated tau spreading accelerates may depend on individual risk factors, such as apolipoprotein E ε4 (ApoE4) carriership, which is linked to faster disease progression; however, the association of ApoE4 with amyloid-related tau spreading is unclear. Objective: To assess if ApoE4 carriers show accelerated amyloid-related tau spreading and propose amyloid positron emission tomography (PET) thresholds at which tau spreading accelerates in ApoE4 carriers vs noncarriers. Design, Setting, and Participants: This cohort study including combined ApoE genotyping, amyloid PET, and longitudinal tau PET from 2 independent samples: the Alzheimer's Disease Neuroimaging Initiative (ADNI; n = 237; collected from April 2015 to August 2022) and Avid-A05 (n = 130; collected from December 2013 to July 2017) with a mean (SD) tau PET follow-up time of 1.9 (0.96) years in ADNI and 1.4 (0.23) years in Avid-A05. ADNI is an observational multicenter Alzheimer disease neuroimaging initiative and Avid-A05 an observational clinical trial. Participants classified as cognitively normal (152 in ADNI and 77 in Avid-A05) or mildly cognitively impaired (107 in ADNI and 53 in Avid-A05) were selected based on ApoE genotyping, amyloid-PET, and longitudinal tau PET data availability. Participants with ApoE ε2/ε4 genotype or classified as having dementia were excluded. Resting-state functional magnetic resonance imaging connectivity templates were based on 42 healthy participants in ADNI. Main Outcomes and Measures: Mediation of amyloid PET on the association between ApoE4 status and subsequent tau PET increase through Braak stage regions and interaction between ApoE4 status and amyloid PET with annual tau PET increase through Braak stage regions and connectivity-based spreading stages (tau epicenter connectivity ranked regions). Results: The mean (SD) age was 73.9 (7.35) years among the 237 ADNI participants and 70.2 (9.7) years among the 130 Avid-A05 participants. A total of 107 individuals in ADNI (45.1%) and 45 in Avid-A05 (34.6%) were ApoE4 carriers. Across both samples, we found that higher amyloid PET-mediated ApoE4-related tau PET increased globally (ADNI b, 0.15; 95% CI, 0.05-0.28; P = .001 and Avid-A05 b, 0.33; 95% CI, 0.14-0.54; P < .001) and in earlier Braak regions. Further, we found a significant association between ApoE4 status by amyloid PET interaction and annual tau PET increases consistently through early Braak- and connectivity-based stages where amyloid-related tau accumulation was accelerated in ApoE4carriers vs noncarriers at lower centiloid thresholds, corrected for age and sex. Conclusions and Relevance: The findings in this study indicate that amyloid-related tau accumulation was accelerated in ApoE4 carriers at lower amyloid levels, suggesting that ApoE4 may facilitate earlier amyloid-driven tau spreading across connected brain regions. Possible therapeutic implications might be further investigated to determine when best to prevent tau spreading and thus cognitive decline depending on ApoE4 status.


Asunto(s)
Enfermedad de Alzheimer , Disfunción Cognitiva , Humanos , Anciano , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/complicaciones , Apolipoproteína E4/genética , Proteínas tau/metabolismo , Anticuerpos Monoclonales , Estudios de Cohortes , Péptidos beta-Amiloides/metabolismo , Amiloide , Apolipoproteínas E/genética , Encéfalo/patología , Tomografía de Emisión de Positrones , Disfunción Cognitiva/patología , Genotipo
4.
Comput Methods Programs Biomed ; 242: 107812, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37757566

RESUMEN

BACKGROUND: Magnetic resonance imaging (MRI), digital pathology imaging (PATH), demographics, and IDH mutation status predict overall survival (OS) in glioma. Identifying and characterizing predictive features in the different modalities may improve OS prediction accuracy. PURPOSE: To evaluate the OS prediction accuracy of combinations of prognostic markers in glioma patients. MATERIALS AND METHODS: Multi-contrast MRI, comprising T1-weighted, T1-weighted post-contrast, T2-weighted, T2 fluid-attenuated-inversion-recovery, and pathology images from glioma patients (n = 160) were retrospectively collected (1983-2008) from TCGA alongside age and sex. Phenotypic profiling of tumors was performed by quantifying the radiographic and histopathologic descriptors extracted from the delineated region-of-interest in MRI and PATH images. A Cox proportional hazard model was trained with the MRI and PATH features, IDH mutation status, and basic demographic variables (age and sex) to predict OS. The performance was evaluated in a split-train-test configuration using the concordance-index, computed between the predicted risk score and observed OS. RESULTS: The average age of patients was 51.2years (women: n = 77, age-range=18-84years; men: n = 83, age-range=21-80years). The median OS of the participants was 494.5 (range,3-4752), 481 (range,7-4752), and 524.5 days (range,3-2869), respectively, in complete dataset, training, and test datasets. The addition of MRI or PATH features improved prediction of OS when compared to models based on age, sex, and mutation status alone or their combination (p < 0.001). The full multi-omics model integrated MRI, PATH, clinical, and genetic profiles and predicted the OS best (c-index= 0.87). CONCLUSION: The combination of imaging, genetic, and clinical profiles leads to a more accurate prognosis than the clinical and/or mutation status.


Asunto(s)
Neoplasias Encefálicas , Glioma , Masculino , Humanos , Femenino , Adolescente , Adulto Joven , Adulto , Persona de Mediana Edad , Anciano , Anciano de 80 o más Años , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/genética , Estudios Retrospectivos , Isocitrato Deshidrogenasa/genética , Glioma/diagnóstico por imagen , Glioma/genética , Imagen por Resonancia Magnética/métodos , Fenotipo , Mutación , Demografía
5.
Cancers (Basel) ; 15(15)2023 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-37568655

RESUMEN

The use of multiparametric magnetic resonance imaging (mpMRI) has become a common technique used in guiding biopsy and developing treatment plans for prostate lesions. While this technique is effective, non-invasive methods such as radiomics have gained popularity for extracting imaging features to develop predictive models for clinical tasks. The aim is to minimize invasive processes for improved management of prostate cancer (PCa). This study reviews recent research progress in MRI-based radiomics for PCa, including the radiomics pipeline and potential factors affecting personalized diagnosis. The integration of artificial intelligence (AI) with medical imaging is also discussed, in line with the development trend of radiogenomics and multi-omics. The survey highlights the need for more data from multiple institutions to avoid bias and generalize the predictive model. The AI-based radiomics model is considered a promising clinical tool with good prospects for application.

6.
J Neuroradiol ; 2023 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-37652263

RESUMEN

PURPOSE: To determine if machine learning (ML) or deep learning (DL) pipelines perform better in AI-based three-class classification of glioblastoma (GBM), intracranial metastatic disease (IMD) and primary CNS lymphoma (PCNSL). METHODOLOGY: Retrospective analysis included 502 cases for training (208 GBM, 67 PCNSL and 227 IMD), with external validation on 86 cases (27:27:32). Multiparametric MRI images (T1W, T2W, FLAIR, DWI and T1-CE) were co-registered, resampled, denoised and intensity normalized, followed by semiautomatic 3D segmentation of the enhancing tumor (ET) and peritumoral region (PTR). Model performance was assessed using several ML pipelines and 3D-convolutional neural networks (3D-CNN) using sequence specific masks, as well as combination of masks. All pipelines were trained and evaluated with 5-fold nested cross-validation on internal data followed by external validation using multi-class AUC. RESULTS: Two ML models achieved similar performance on test set, one using T2-ET and T2-PTR masks (AUC: 0.885, 95% CI: [0.816, 0.935] and another using T1-CE-ET and FLAIR-PTR mask (AUC: 0.878, CI: [0.804, 0.930]). The best performing DL models achieved an AUC of 0.854, (CI [0.774, 0.914]) on external data using T1-CE-ET and T2-PTR masks, followed by model derived from T1-CE-ET, ADC-ET and FLAIR-PTR masks (AUC: 0.851, CI [0.772, 0.909]). CONCLUSION: Both ML and DL derived pipelines achieved similar performance. T1-CE mask was used in three of the top four overall models. Additionally, all four models had some mask derived from PTR, either T2WI or FLAIR.

7.
Sci Data ; 9(1): 453, 2022 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-35906241

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Adulto , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/fisiopatología , Genómica , Glioblastoma/diagnóstico por imagen , Glioblastoma/genética , Glioblastoma/fisiopatología , Humanos , Imagen por Resonancia Magnética , Pronóstico
8.
Appl Sci (Basel) ; 11(16)2021 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-34621541

RESUMEN

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.

9.
Radiol Imaging Cancer ; 3(4): e200108, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34296969

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas , Glioma , Neoplasias Encefálicas/diagnóstico por imagen , Glioma/diagnóstico por imagen , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Máquina de Vectores de Soporte
10.
Cancers (Basel) ; 13(3)2021 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-33535569

RESUMEN

The management of prostate cancer (PCa) is dependent on biomarkers of biological aggression. This includes an invasive biopsy to facilitate a histopathological assessment of the tumor's grade. This review explores the technical processes of applying magnetic resonance imaging based radiomic models to the evaluation of PCa. By exploring how a deep radiomics approach further optimizes the prediction of a PCa's grade group, it will be clear how this integration of artificial intelligence mitigates existing major technological challenges faced by a traditional radiomic model: image acquisition, small data sets, image processing, labeling/segmentation, informative features, predicting molecular features and incorporating predictive models. Other potential impacts of artificial intelligence on the personalized treatment of PCa will also be discussed. The role of deep radiomics analysis-a deep texture analysis, which extracts features from convolutional neural networks layers, will be highlighted. Existing clinical work and upcoming clinical trials will be reviewed, directing investigators to pertinent future directions in the field. For future progress to result in clinical translation, the field will likely require multi-institutional collaboration in producing prospectively populated and expertly labeled imaging libraries.

11.
Brainlesion ; 11993: 380-394, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32754723

RESUMEN

The purpose of this manuscript is to provide an overview of the technical specifications and architecture of the Cancer imaging Phenomics Toolkit (CaPTk www.cbica.upenn.edu/captk), a cross-platform, open-source, easy-to-use, and extensible software platform for analyzing 2D and 3D images, currently focusing on radiographic scans of brain, breast, and lung cancer. The primary aim of this platform is to enable swift and efficient translation of cutting-edge academic research into clinically useful tools relating to clinical quantification, analysis, predictive modeling, decision-making, and reporting workflow. CaPTk builds upon established open-source software toolkits, such as the Insight Toolkit (ITK) and OpenCV, to bring together advanced computational functionality. This functionality describes specialized, as well as general-purpose, image analysis algorithms developed during active multi-disciplinary collaborative research studies to address real clinical requirements. The target audience of CaPTk consists of both computational scientists and clinical experts. For the former it provides i) an efficient image viewer offering the ability of integrating new algorithms, and ii) a library of readily-available clinically-relevant algorithms, allowing batch-processing of multiple subjects. For the latter it facilitates the use of complex algorithms for clinically-relevant studies through a user-friendly interface, eliminating the prerequisite of a substantial computational background. CaPTk's long-term goal is to provide widely-used technology to make use of advanced quantitative imaging analytics in cancer prediction, diagnosis and prognosis, leading toward a better understanding of the biological mechanisms of cancer development.

12.
Neuroimage ; 220: 117081, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32603860

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas/diagnóstico por imagen , Encéfalo/diagnóstico por imagen , Glioma/diagnóstico por imagen , Procesamiento de Imagen Asistido por Computador/métodos , Imagen por Resonancia Magnética/métodos , Bases de Datos Factuales , Aprendizaje Profundo , Humanos , Estudios Retrospectivos
13.
J Med Imaging (Bellingham) ; 7(3): 031505, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32566694

RESUMEN

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.

15.
JCO Clin Cancer Inform ; 4: 234-244, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32191542

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas/mortalidad , Glioblastoma/mortalidad , Interpretación de Imagen Asistida por Computador/métodos , Imágenes de Resonancia Magnética Multiparamétrica/métodos , Recurrencia Local de Neoplasia/mortalidad , Fenómica/métodos , Programas Informáticos , Adulto , Anciano , Anciano de 80 o más Años , Algoritmos , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/cirugía , Femenino , Glioblastoma/metabolismo , Glioblastoma/patología , Glioblastoma/cirugía , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/cirugía , Supervivencia sin Progresión , Curva ROC , Estudios Retrospectivos , Tasa de Supervivencia , Adulto Joven
16.
Cancers (Basel) ; 12(3)2020 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-32131409

RESUMEN

Cancer pathology reflects disease progression (or regression) and associated molecular characteristics, and provides rich phenotypic information that is predictive of cancer grade and has potential implications in treatment planning and prognosis. According to the remarkable performance of computational approaches in the digital pathology domain, we hypothesized that machine learning can help to distinguish low-grade gliomas (LGG) from high-grade gliomas (HGG) by exploiting the rich phenotypic information that reflects the microvascular proliferation level, mitotic activity, presence of necrosis, and nuclear atypia present in digital pathology images. A set of 735 whole-slide digital pathology images of glioma patients (median age: 49.65 years, male: 427, female: 308, median survival: 761.26 days) were obtained from TCGA. Sub-images that contained a viable tumor area, showing sufficient histologic characteristics, and that did not have any staining artifact were extracted. Several clinical measures and imaging features, including conventional (intensity, morphology) and advanced textures features (gray-level co-occurrence matrix and gray-level run-length matrix), extracted from the sub-images were further used for training the support vector machine model with linear configuration. We sought to evaluate the combined effect of conventional imaging, clinical, and texture features by assessing the predictive value of each feature type and their combinations through a predictive classifier. The texture features were successfully validated on the glioma patients in 10-fold cross-validation (accuracy = 75.12%, AUC = 0.652). The addition of texture features to clinical and conventional imaging features improved grade prediction compared to the models trained on clinical and conventional imaging features alone (p = 0.045 and p = 0.032 for conventional imaging features and texture features, respectively). The integration of imaging, texture, and clinical features yielded a significant improvement in accuracy, supporting the synergistic value of these features in the predictive model. The findings suggest that the texture features, when combined with conventional imaging and clinical markers, may provide an objective, accurate, and integrated prediction of glioma grades. The proposed digital pathology imaging-based marker may help to (i) stratify patients into clinical trials, (ii) select patients for targeted therapies, and (iii) personalize treatment planning on an individual person basis.

17.
Cancer ; 126(11): 2625-2636, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32129893

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Aprendizaje Automático , Imagen por Resonancia Magnética/métodos , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor , Neoplasias Encefálicas/diagnóstico por imagen , Progresión de la Enfermedad , Femenino , Glioblastoma/diagnóstico por imagen , Humanos , Masculino , Persona de Mediana Edad
18.
Artículo en Inglés | MEDLINE | ID: mdl-33746333

RESUMEN

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.
Artículo en Inglés | MEDLINE | ID: mdl-33521638

RESUMEN

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.
Cancers (Basel) ; 11(11)2019 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-31683818

RESUMEN

Distinguishing benign from malignant disease is a primary challenge for colon histopathologists. Current clinical methods rely on qualitative visual analysis of features such as glandular architecture and size that exist on a continuum from benign to malignant. Consequently, discordance between histopathologists is common. To provide more reliable analysis of colon specimens, we propose an end-to-end computational pathology pipeline that encompasses gland segmentation, cancer detection, and then further breaking down the malignant samples into different cancer grades. We propose a multi-step gland segmentation method, which models tissue components as ellipsoids. For cancer detection/grading, we encode cellular morphology, spatial architectural patterns of glands, and texture by extracting multi-scale features: (i) Gland-based: extracted from individual glands, (ii) local-patch-based: computed from randomly-selected image patches, and (iii) image-based: extracted from images, and employ a hierarchical ensemble-classification method. Using two datasets (Rawalpindi Medical College (RMC), n = 174 and gland segmentation (GlaS), n = 165) with three cancer grades, our method reliably delineated gland regions (RMC = 87.5%, GlaS = 88.4%), detected the presence of malignancy (RMC = 97.6%, GlaS = 98.3%), and predicted tumor grade (RMC = 98.6%, GlaS = 98.6%). Training the model using one dataset and testing it on the other showed strong concordance in cancer detection (Train RMC - Test GlaS = 94.5%, Train GlaS - Test RMC = 93.7%) and grading (Train RMC - Test GlaS = 95%, Train GlaS - Test RMC = 95%) suggesting that the model will be applicable across institutions. With further prospective validation, the techniques demonstrated here may provide a reproducible and easily accessible method to standardize analysis of colon cancer specimens.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...