Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Radiology ; 310(2): e231319, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38319168

RESUMO

Filters are commonly used to enhance specific structures and patterns in images, such as vessels or peritumoral regions, to enable clinical insights beyond the visible image using radiomics. However, their lack of standardization restricts reproducibility and clinical translation of radiomics decision support tools. In this special report, teams of researchers who developed radiomics software participated in a three-phase study (September 2020 to December 2022) to establish a standardized set of filters. The first two phases focused on finding reference filtered images and reference feature values for commonly used convolutional filters: mean, Laplacian of Gaussian, Laws and Gabor kernels, separable and nonseparable wavelets (including decomposed forms), and Riesz transformations. In the first phase, 15 teams used digital phantoms to establish 33 reference filtered images of 36 filter configurations. In phase 2, 11 teams used a chest CT image to derive reference values for 323 of 396 features computed from filtered images using 22 filter and image processing configurations. Reference filtered images and feature values for Riesz transformations were not established. Reproducibility of standardized convolutional filters was validated on a public data set of multimodal imaging (CT, fluorodeoxyglucose PET, and T1-weighted MRI) in 51 patients with soft-tissue sarcoma. At validation, reproducibility of 486 features computed from filtered images using nine configurations × three imaging modalities was assessed using the lower bounds of 95% CIs of intraclass correlation coefficients. Out of 486 features, 458 were found to be reproducible across nine teams with lower bounds of 95% CIs of intraclass correlation coefficients greater than 0.75. In conclusion, eight filter types were standardized with reference filtered images and reference feature values for verifying and calibrating radiomics software packages. A web-based tool is available for compliance checking.


Assuntos
Processamento de Imagem Assistida por Computador , Radiômica , Humanos , Reprodutibilidade dos Testes , Biomarcadores , Imagem Multimodal
2.
Brainlesion ; 13769: 68-79, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37928819

RESUMO

Convolutional neural networks (CNNs) have shown promising performance in various 2D computer vision tasks due to availability of large amounts of 2D training data. Contrarily, medical imaging deals with 3D data and usually lacks the equivalent extent and diversity of data, for developing AI models. Transfer learning provides the means to use models trained for one application as a starting point to another application. In this work, we leverage 2D pre-trained models as a starting point in 3D medical applications by exploring the concept of Axial-Coronal-Sagittal (ACS) convolutions. We have incorporated ACS as an alternative of native 3D convolutions in the Generally Nuanced Deep Learning Framework (GaNDLF), providing various well-established and state-of-the-art network architectures with the availability of pre-trained encoders from 2D data. Results of our experimental evaluation on 3D MRI data of brain tumor patients for i) tumor segmentation and ii) radiogenomic classification, show model size reduction by ~22% and improvement in validation accuracy by ~33%. Our findings support the advantage of ACS convolutions in pre-trained 2D CNNs over 3D CNN without pre-training, for 3D segmentation and classification tasks, democratizing existing models trained in datasets of unprecedented size and showing promise in the field of healthcare.

3.
ArXiv ; 2023 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-37332562

RESUMO

Software is vital for the advancement of biology and medicine. Through analysis of usage and impact metrics of software, developers can help determine user and community engagement. These metrics can be used to justify additional funding, encourage additional use, and identify unanticipated use cases. Such analyses can help define improvement areas and assist with managing project resources. However, there are challenges associated with assessing usage and impact, many of which vary widely depending on the type of software being evaluated. These challenges involve issues of distorted, exaggerated, understated, or misleading metrics, as well as ethical and security concerns. More attention to the nuances, challenges, and considerations involved in capturing impact across the diverse spectrum of biological software is needed. Furthermore, some tools may be especially beneficial to a small audience, yet may not have comparatively compelling metrics of high usage. Although some principles are generally applicable, there is not a single perfect metric or approach to effectively evaluate a software tool's impact, as this depends on aspects unique to each tool, how it is used, and how one wishes to evaluate engagement. We propose more broadly applicable guidelines (such as infrastructure that supports the usage of software and the collection of metrics about usage), as well as strategies for various types of software and resources. We also highlight outstanding issues in the field regarding how communities measure or evaluate software impact. To gain a deeper understanding of the issues hindering software evaluations, as well as to determine what appears to be helpful, we performed a survey of participants involved with scientific software projects for the Informatics Technology for Cancer Research (ITCR) program funded by the National Cancer Institute (NCI). We also investigated software among this scientific community and others to assess how often infrastructure that supports such evaluations is implemented and how this impacts rates of papers describing usage of the software. We find that although developers recognize the utility of analyzing data related to the impact or usage of their software, they struggle to find the time or funding to support such analyses. We also find that infrastructure such as social media presence, more in-depth documentation, the presence of software health metrics, and clear information on how to contact developers seem to be associated with increased usage rates. Our findings can help scientific software developers make the most out of the evaluations of their software so that they can more fully benefit from such assessments.

5.
Nat Mach Intell ; 5(7): 799-810, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38706981

RESUMO

Medical artificial intelligence (AI) has tremendous potential to advance healthcare by supporting and contributing to the evidence-based practice of medicine, personalizing patient treatment, reducing costs, and improving both healthcare provider and patient experience. Unlocking this potential requires systematic, quantitative evaluation of the performance of medical AI models on large-scale, heterogeneous data capturing diverse patient populations. Here, to meet this need, we introduce MedPerf, an open platform for benchmarking AI models in the medical domain. MedPerf focuses on enabling federated evaluation of AI models, by securely distributing them to different facilities, such as healthcare organizations. This process of bringing the model to the data empowers each facility to assess and verify the performance of AI models in an efficient and human-supervised process, while prioritizing privacy. We describe the current challenges healthcare and AI communities face, the need for an open platform, the design philosophy of MedPerf, its current implementation status and real-world deployment, our roadmap and, importantly, the use of MedPerf with multiple international institutions within cloud-based technology and on-premises scenarios. Finally, we welcome new contributions by researchers and organizations to further strengthen MedPerf as an open benchmarking platform.

6.
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
7.
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.

8.
Phys Med Biol ; 67(21)2022 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-36198326

RESUMO

Objective.Federated learning (FL) is a computational paradigm that enables organizations to collaborate on machine learning (ML) and deep learning (DL) projects without sharing sensitive data, such as patient records, financial data, or classified secrets.Approach.Open federated learning (OpenFL) framework is an open-source python-based tool for training ML/DL algorithms using the data-private collaborative learning paradigm of FL, irrespective of the use case. OpenFL works with training pipelines built with both TensorFlow and PyTorch, and can be easily extended to other ML and DL frameworks.Main results.In this manuscript, we present OpenFL and summarize its motivation and development characteristics, with the intention of facilitating its application to existing ML/DL model training in a production environment. We further provide recommendations to secure a federation using trusted execution environments to ensure explicit model security and integrity, as well as maintain data confidentiality. Finally, we describe the first real-world healthcare federations that use the OpenFL library, and highlight how it can be applied to other non-healthcare use cases.Significance.The OpenFL library is designed for real world scalability, trusted execution, and also prioritizes easy migration of centralized ML models into a federated training pipeline. Although OpenFL's initial use case was in healthcare, it is applicable beyond this domain and is now reaching wider adoption both in research and production settings. The tool is open-sourced atgithub.com/intel/openfl.


Assuntos
Algoritmos , Aprendizado de Máquina , Humanos
9.
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
10.
Sci Data ; 9(1): 440, 2022 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-35871247

RESUMO

Breast cancer is one of the most pervasive forms of cancer and its inherent intra- and inter-tumor heterogeneity contributes towards its poor prognosis. Multiple studies have reported results from either private institutional data or publicly available datasets. However, current public datasets are limited in terms of having consistency in: a) data quality, b) quality of expert annotation of pathology, and c) availability of baseline results from computational algorithms. To address these limitations, here we propose the enhancement of the I-SPY1 data collection, with uniformly curated data, tumor annotations, and quantitative imaging features. Specifically, the proposed dataset includes a) uniformly processed scans that are harmonized to match intensity and spatial characteristics, facilitating immediate use in computational studies, b) computationally-generated and manually-revised expert annotations of tumor regions, as well as c) a comprehensive set of quantitative imaging (also known as radiomic) features corresponding to the tumor regions. This collection describes our contribution towards repeatable, reproducible, and comparative quantitative studies leading to new predictive, prognostic, and diagnostic assessments.


Assuntos
Neoplasias da Mama , Algoritmos , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/patologia , Feminino , Humanos , Imageamento por Ressonância Magnética
11.
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
12.
Artigo em Inglês | MEDLINE | ID: mdl-35465060

RESUMO

Diabetic foot ulceration (DFU) is a serious complication of diabetes, and a major challenge for healthcare systems around the world. Further infection and ischemia in DFU can significantly prolong treatment and often result in limb amputation, with more severe cases resulting in terminal illness. Thus, early identification and regular monitoring is necessary to improve care, and reduce the burden on healthcare systems. With that in mind, this study attempts to address the problem of infection and ischemia classification in diabetic food ulcers, in four distinct classes. We have evaluated a series of VGG architectures with different layers, following numerous training strategies, including k-fold cross validation, data pre-processing options, augmentation techniques, and weighted loss calculations. In favor of transparency and reproducibility, we make all the implementations available through the Generally Nuanced Deep Learning Framework (GaNDLF, github.com/CBICA/GaNDLF. Our best model was evaluated during the DFU Challenge 2021, and was ranked 2nd, 5th, and 7th based on the macro-averaged AUC (area under the curve), macro-averaged F1 score, and macro-averaged recall metrics, respectively. Our findings support that current state-of-the-art architectures provide good results for the DFU image classification task, and further experimentation is required to study the effects of pre-processing and augmentation strategies.

13.
IEEE Open J Eng Med Biol ; 3: 218-226, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36860498

RESUMO

Histopathologic evaluation of Hematoxylin & Eosin (H&E) stained slides is essential for disease diagnosis, revealing tissue morphology, structure, and cellular composition. Variations in staining protocols and equipment result in images with color nonconformity. Although pathologists compensate for color variations, these disparities introduce inaccuracies in computational whole slide image (WSI) analysis, accentuating data domain shift and degrading generalization. Current state-of-the-art normalization methods employ a single WSI as reference, but selecting a single WSI representative of a complete WSI-cohort is infeasible, inadvertently introducing normalization bias. We seek the optimal number of slides to construct a more representative reference based on composite/aggregate of multiple H&E density histograms and stain-vectors, obtained from a randomly selected WSI population (WSI-Cohort-Subset). We utilized 1,864 IvyGAP WSIs as a WSI-cohort, and built 200 WSI-Cohort-Subsets varying in size (from 1 to 200 WSI-pairs) using randomly selected WSIs. The WSI-pairs' mean Wasserstein Distances and WSI-Cohort-Subsets' standard deviations were calculated. The Pareto Principle defined the optimal WSI-Cohort-Subset size. The WSI-cohort underwent structure-preserving color normalization using the optimal WSI-Cohort-Subset histogram and stain-vector aggregates. Numerous normalization permutations support WSI-Cohort-Subset aggregates as representative of a WSI-cohort through WSI-cohort CIELAB color space swift convergence, as a result of the law of large numbers and shown as a power law distribution. We show normalization at the optimal (Pareto Principle) WSI-Cohort-Subset size and corresponding CIELAB convergence: a) Quantitatively, using 500 WSI-cohorts; b) Quantitatively, using 8,100 WSI-regions; c) Qualitatively, using 30 cellular tumor normalization permutations. Aggregate-based stain normalization may contribute in increasing computational pathology robustness, reproducibility, and integrity.

14.
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.

15.
Brainlesion ; 12658: 157-167, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34514469

RESUMO

Glioblastoma ( GBM ) is arguably the most aggressive, infiltrative, and heterogeneous type of adult brain tumor. Biophysical modeling of GBM growth has contributed to more informed clinical decision-making. However, deploying a biophysical model to a clinical environment is challenging since underlying computations are quite expensive and can take several hours using existing technologies. Here we present a scheme to accelerate the computation. In particular, we present a deep learning ( DL )-based logistic regression model to estimate the GBM's biophysical growth in seconds. This growth is defined by three tumor-specific parameters: 1) a diffusion coefficient in white matter ( Dw ), which prescribes the rate of infiltration of tumor cells in white matter, 2) a mass-effect parameter ( Mp ), which defines the average tumor expansion, and 3) the estimated time ( T ) in number of days that the tumor has been growing. Preoperative structural multi-parametric MRI ( mpMRI ) scans from n = 135 subjects of the TCGA-GBM imaging collection are used to quantitatively evaluate our approach. We consider the mpMRI intensities within the region defined by the abnormal FLAIR signal envelope for training one DL model for each of the tumor-specific growth parameters. We train and validate the DL-based predictions against parameters derived from biophysical inversion models. The average Pearson correlation coefficients between our DL-based estimations and the biophysical parameters are 0.85 for Dw, 0.90 for Mp, and 0.94 for T, respectively. This study unlocks the power of tumor-specific parameters from biophysical tumor growth estimation. It paves the way towards their clinical translation and opens the door for leveraging advanced radiomic descriptors in future studies by means of a significantly faster parameter reconstruction compared to biophysical growth modeling approaches.

16.
Appl Sci (Basel) ; 11(4)2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34290888

RESUMO

Histopathologic assessment routinely provides rich microscopic information about tissue structure and disease process. However, the sections used are very thin, and essentially capture only 2D representations of a certain tissue sample. Accurate and robust alignment of sequentially cut 2D slices should contribute to more comprehensive assessment accounting for surrounding 3D information. Towards this end, we here propose a two-step diffeomorphic registration approach that aligns differently stained histology slides to each other, starting with an initial affine step followed by estimating a deformation field. It was quantitatively evaluated on ample (n = 481) and diverse data from the automatic non-rigid histological image registration challenge, where it was awarded the second rank. The obtained results demonstrate the ability of the proposed approach to robustly (average robustness = 0.9898) and accurately (average relative target registration error = 0.2%) align differently stained histology slices of various anatomical sites while maintaining reasonable computational efficiency (<1 min per registration). The method was developed by adapting a general-purpose registration algorithm designed for 3D radiographic scans and achieved consistently accurate results for aligning high-resolution 2D histologic images. Accurate alignment of histologic images can contribute to a better understanding of the spatial arrangement and growth patterns of cells, vessels, matrix, nerves, and immune cell interactions.

17.
Front Neurosci ; 15: 752780, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35035351

RESUMO

A multitude of image-based machine learning segmentation and classification algorithms has recently been proposed, offering diagnostic decision support for the identification and characterization of glioma, Covid-19 and many other diseases. Even though these algorithms often outperform human experts in segmentation tasks, their limited reliability, and in particular the inability to detect failure cases, has hindered translation into clinical practice. To address this major shortcoming, we propose an unsupervised quality estimation method for segmentation ensembles. Our primitive solution examines discord in binary segmentation maps to automatically flag segmentation results that are particularly error-prone and therefore require special assessment by human readers. We validate our method both on segmentation of brain glioma in multi-modal magnetic resonance - and of lung lesions in computer tomography images. Additionally, our method provides an adaptive prioritization mechanism to maximize efficacy in use of human expert time by enabling radiologists to focus on the most difficult, yet important cases while maintaining full diagnostic autonomy. Our method offers an intuitive and reliable uncertainty estimation from segmentation ensembles and thereby closes an important gap toward successful translation of automatic segmentation into clinical routine.

18.
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
19.
Brainlesion ; 11993: 380-394, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32754723

RESUMO

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.

20.
Sci Rep ; 10(1): 12598, 2020 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-32724046

RESUMO

Several studies underscore the potential of deep learning in identifying complex patterns, leading to diagnostic and prognostic biomarkers. Identifying sufficiently large and diverse datasets, required for training, is a significant challenge in medicine and can rarely be found in individual institutions. Multi-institutional collaborations based on centrally-shared patient data face privacy and ownership challenges. Federated learning is a novel paradigm for data-private multi-institutional collaborations, where model-learning leverages all available data without sharing data between institutions, by distributing the model-training to the data-owners and aggregating their results. We show that federated learning among 10 institutions results in models reaching 99% of the model quality achieved with centralized data, and evaluate generalizability on data from institutions outside the federation. We further investigate the effects of data distribution across collaborating institutions on model quality and learning patterns, indicating that increased access to data through data private multi-institutional collaborations can benefit model quality more than the errors introduced by the collaborative method. Finally, we compare with other collaborative-learning approaches demonstrating the superiority of federated learning, and discuss practical implementation considerations. Clinical adoption of federated learning is expected to lead to models trained on datasets of unprecedented size, hence have a catalytic impact towards precision/personalized medicine.


Assuntos
Disseminação de Informação , Relações Interinstitucionais , Aprendizagem , Medicina , Pacientes , Privacidade , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...