Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
IEEE Winter Conf Appl Comput Vis ; 2024: 5170-5179, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38808304

RESUMO

To achieve high-quality results, diffusion models must be trained on large datasets. This can be notably prohibitive for models in specialized domains, such as computational pathology. Conditioning on labeled data is known to help in data-efficient model training. Therefore, histopathology reports, which are rich in valuable clinical information, are an ideal choice as guidance for a histopathology generative model. In this paper, we introduce PathLDM, the first text-conditioned Latent Diffusion Model tailored for generating high-quality histopathology images. Leveraging the rich contextual information provided by pathology text reports, our approach fuses image and textual data to enhance the generation process. By utilizing GPT's capabilities to distill and summarize complex text reports, we establish an effective conditioning mechanism. Through strategic conditioning and necessary architectural enhancements, we achieved a SoTA FID score of 7.64 for text-to-image generation on the TCGA-BRCA dataset, significantly outperforming the closest text-conditioned competitor with FID 30.1.

2.
Adv Radiat Oncol ; 9(5): 101457, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38550363

RESUMO

Purpose: Stereotactic radiosurgery/radiation therapy (SRS/SRT) increasingly has been used to treat brain metastases. However, the development of distant brain metastases (DBMs) in the untreated brain remains a serious complication. We sought to develop a spatially aware radiomic signature to model the time-to-DBM development in a cohort of patients leveraging pretreatment magnetic resonance imaging (MRI) and radiation therapy treatment planning data including radiation dose distribution maps. Methods and Materials: We retrospectively analyzed a cohort of 105 patients with brain metastases treated by SRS/SRT with pretreatment multiparametric MRI (T1, T1 postcontrast, T2, fluid-attenuated inversion recovery). Three-dimensional radiomic features were extracted from each MRI sequence within 5 isodose regions of interest (ROIs) identified via radiation dose distribution maps and gross target volume (GTV) contours. Clinical features including patient performance status, number of lesions treated, tumor volume, and tumor stage were collected to serve as a baseline for comparison. Cox proportional hazards (CPH) modeling and Kaplan-Meier analysis were used to model time-to-DBM development. Results: CPH models trained using radiomic features achieved a mean concordance index (c-index) of 0.63 (standard deviation [SD], 0.08) compared with a c-index of 0.49 (SD, 0.09) for CPH models trained using clinical factors. A CPH model trained using both radiomic and clinical features achieved a c-index of 0.69 (SD, 0.08). The identified radiomic signature was able to stratify patients into distinct risk groups with statistically significant differences (P = .00007) in time-to-DBM development as measured by log-rank test. Clinical features were unable to do the same. Radiomic features from the peritumoral 50% to 75% isodose ROI and GTV region were most predictive of DBM development. Conclusions: Our results suggest that radiomic features extracted from pretreatment MRI and multiple isodose ROIs can model time-to-DBM development in patients receiving SRS/SRT for brain metastases, outperforming clinical feature baselines. Notably, we believe we are the first to leverage SRS/SRT dose maps for ROI identification and subsequent radiomic analysis of peritumoral and untargeted brain regions using multiparametric MRI. We observed that the peritumoral environment may be implicated in DBM development for SRS/SRT-treated brain metastases. Our preliminary results might enable the identification of patients with predisposition to DBM development and prompt subsequent changes in disease management.

3.
Med Image Anal ; 93: 103070, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38176354

RESUMO

We propose DiRL, a Diversity-inducing Representation Learning technique for histopathology imaging. Self-supervised learning (SSL) techniques, such as contrastive and non-contrastive approaches, have been shown to learn rich and effective representations of digitized tissue samples with limited pathologist supervision. Our analysis of vanilla SSL-pretrained models' attention distribution reveals an insightful observation: sparsity in attention, i.e, models tends to localize most of their attention to some prominent patterns in the image. Although attention sparsity can be beneficial in natural images due to these prominent patterns being the object of interest itself, this can be sub-optimal in digital pathology; this is because, unlike natural images, digital pathology scans are not object-centric, but rather a complex phenotype of various spatially intermixed biological components. Inadequate diversification of attention in these complex images could result in crucial information loss. To address this, we leverage cell segmentation to densely extract multiple histopathology-specific representations, and then propose a prior-guided dense pretext task, designed to match the multiple corresponding representations between the views. Through this, the model learns to attend to various components more closely and evenly, thus inducing adequate diversification in attention for capturing context-rich representations. Through quantitative and qualitative analysis on multiple tasks across cancer types, we demonstrate the efficacy of our method and observe that the attention is more globally distributed.


Assuntos
Processamento de Imagem Assistida por Computador , Aprendizado de Máquina , Patologia , Humanos , Fenótipo , Patologia/métodos
4.
Proc IEEE Int Conf Comput Vis ; 2023: 21358-21368, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38737337

RESUMO

In medical vision, different imaging modalities provide complementary information. However, in practice, not all modalities may be available during inference or even training. Previous approaches, e.g., knowledge distillation or image synthesis, often assume the availability of full modalities for all subjects during training; this is unrealistic and impractical due to the variability in data collection across sites. We propose a novel approach to learn enhanced modality-agnostic representations by employing a meta-learning strategy in training, even when only limited full modality samples are available. Meta-learning enhances partial modality representations to full modality representations by meta-training on partial modality data and meta-testing on limited full modality samples. Additionally, we co-supervise this feature enrichment by introducing an auxiliary adversarial learning branch. More specifically, a missing modality detector is used as a discriminator to mimic the full modality setting. Our segmentation framework significantly outperforms state-of-the-art brain tumor segmentation techniques in missing modality scenarios.

5.
Sci Adv ; 8(47): eabq4609, 2022 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-36427313

RESUMO

Tumor vasculature is a key component of the tumor microenvironment that can influence tumor behavior and therapeutic resistance. We present a new imaging biomarker, quantitative vessel tortuosity (QVT), and evaluate its association with response and survival in patients with non-small cell lung cancer (NSCLC) treated with immune checkpoint inhibitor (ICI) therapies. A total of 507 cases were used to evaluate different aspects of the QVT biomarkers. QVT features were extracted from computed tomography imaging of patients before and after ICI therapy to capture the tortuosity, curvature, density, and branching statistics of the nodule vasculature. Our results showed that QVT features were prognostic of OS (HR = 3.14, 0.95% CI = 1.2 to 9.68, P = 0.0006, C-index = 0.61) and could predict ICI response with AUCs of 0.66, 0.61, and 0.67 on three validation sets. Our study shows that QVT imaging biomarker could potentially aid in predicting and monitoring response to ICI in patients with NSCLC.

6.
NPJ Precis Oncol ; 6(1): 33, 2022 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-35661148

RESUMO

Despite known histological, biological, and clinical differences between lung adenocarcinoma (LUAD) and squamous cell carcinoma (LUSC), relatively little is known about the spatial differences in their corresponding immune contextures. Our study of over 1000 LUAD and LUSC tumors revealed that computationally derived patterns of tumor-infiltrating lymphocytes (TILs) on H&E images were different between LUAD (N = 421) and LUSC (N = 438), with TIL density being prognostic of overall survival in LUAD and spatial arrangement being more prognostically relevant in LUSC. In addition, the LUAD-specific TIL signature was associated with OS in an external validation set of 100 NSCLC treated with more than six different neoadjuvant chemotherapy regimens, and predictive of response to therapy in the clinical trial CA209-057 (n = 303). In LUAD, the prognostic TIL signature was primarily comprised of CD4+ T and CD8+ T cells, whereas in LUSC, the immune patterns were comprised of CD4+ T, CD8+ T, and CD20+ B cells. In both subtypes, prognostic TIL features were associated with transcriptomics-derived immune scores and biological pathways implicated in immune recognition, response, and evasion. Our results suggest the need for histologic subtype-specific TIL-based models for stratifying survival risk and predicting response to therapy. Our findings suggest that predictive models for response to therapy will need to account for the unique morphologic and molecular immune patterns as a function of histologic subtype of NSCLC.

7.
Clin Cancer Res ; 28(20): 4410-4424, 2022 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-35727603

RESUMO

PURPOSE: The tumor-associated vasculature (TAV) differs from healthy blood vessels by its convolutedness, leakiness, and chaotic architecture, and these attributes facilitate the creation of a treatment-resistant tumor microenvironment. Measurable differences in these attributes might also help stratify patients by likely benefit of systemic therapy (e.g., chemotherapy). In this work, we present a new category of computational image-based biomarkers called quantitative tumor-associated vasculature (QuanTAV) features, and demonstrate their ability to predict response and survival across multiple cancer types, imaging modalities, and treatment regimens involving chemotherapy. EXPERIMENTAL DESIGN: We isolated tumor vasculature and extracted mathematical measurements of twistedness and organization from routine pretreatment radiology (CT or contrast-enhanced MRI) of a total of 558 patients, who received one of four first-line chemotherapy-based therapeutic intervention strategies for breast (n = 371) or non-small cell lung cancer (NSCLC, n = 187). RESULTS: Across four chemotherapy-based treatment strategies, classifiers of QuanTAV measurements significantly (P < 0.05) predicted response in held out testing cohorts alone (AUC = 0.63-0.71) and increased AUC by 0.06-0.12 when added to models of significant clinical variables alone. Similarly, we derived QuanTAV risk scores that were prognostic of recurrence-free survival in treatment cohorts who received surgery following chemotherapy for breast cancer [P = 0.0022; HR = 1.25; 95% confidence interval (CI), 1.08-1.44; concordance index (C-index) = 0.66] and chemoradiation for NSCLC (P = 0.039; HR = 1.28; 95% CI, 1.01-1.62; C-index = 0.66). From vessel-based risk scores, we further derived categorical QuanTAV high/low risk groups that were independently prognostic among all treatment groups, including patients with NSCLC who received chemotherapy only (P = 0.034; HR = 2.29; 95% CI, 1.07-4.94; C-index = 0.62). QuanTAV response and risk scores were independent of clinicopathologic risk factors and matched or exceeded models of clinical variables including posttreatment response. CONCLUSIONS: Across these domains, we observed an association of vascular morphology on CT and MRI-as captured by metrics of vessel curvature, torsion, and organizational heterogeneity-and treatment outcome. Our findings suggest the potential of shape and structure of the TAV in developing prognostic and predictive biomarkers for multiple cancers and different treatment strategies.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Biomarcadores , Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Quimiorradioterapia/métodos , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Tomografia Computadorizada por Raios X , Microambiente Tumoral
8.
IEEE Trans Med Imaging ; 41(7): 1764-1777, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35108202

RESUMO

The concept of tumor field effect implies that cancer is a systemic disease with its impact way beyond the visible tumor confines. For instance, in Glioblastoma (GBM), an aggressive brain tumor, the increase in intracranial pressure due to tumor burden often leads to brain herniation and poor outcomes. Our work is based on the rationale that highly aggressive tumors tend to grow uncontrollably, leading to pronounced biomechanical tissue deformations in the normal parenchyma, which when combined with local morphological differences in the tumor confines on MRI scans, will comprehensively capture tumor field effect. Specifically, we present an integrated MRI-based descriptor, radiomic-Deformation and Textural Heterogeneity (r-DepTH). This descriptor comprises measurements of the subtle perturbations in tissue deformations throughout the surrounding normal parenchyma due to mass effect. This involves non-rigidly aligning the patients' MRI scans to a healthy atlas via diffeomorphic registration. The resulting inverse mapping is used to obtain the deformation field magnitudes in the normal parenchyma. These measurements are then combined with a 3D texture descriptor, Co-occurrence of Local Anisotropic Gradient Orientations (COLLAGE), which captures the morphological heterogeneity and infiltration within the tumor confines, on MRI scans. In this work, we extensively evaluated r-DepTH for survival risk-stratification on a total of 207 GBM cases from 3 different cohorts (Cohort 1 ( n1 = 53 ), Cohort 2 ( n2 = 75 ), and Cohort 3 ( n3 = 79 )), where each of these three cohorts was used as a training set for our model separately, and the other two cohorts were used for testing, independently, for each training experiment. When employing Cohort 1 for training, r-DepTH yielded Concordance indices (C-indices) of 0.7 and 0.65, hazard ratios (HR) and Confidence Intervals (CI) of 10 (6 - 19) and 5 (3 - 8) on Cohorts 2 and 3, respectively. Similarly, training on Cohort 2 yielded C-indices of 0.6 and 0.7, HR and CI of 1 (0.7 - 2) and 3 (2 - 5) on Cohorts 1 and 3, respectively. Finally, training on Cohort 3 yielded C-indices of 0.75 and 0.63, HR and CI of 24 (10 - 57) and 12 (6 - 21) on Cohorts 1 and 2, respectively. Our results show that r-DepTH descriptor may serve as a comprehensive and a robust MRI-based prognostic marker of disease aggressiveness and survival in solid tumors.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Anisotropia , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/patologia , Estudos de Coortes , Glioblastoma/diagnóstico por imagem , Glioblastoma/patologia , Humanos , Imageamento por Ressonância Magnética/métodos , Prognóstico
9.
IEEE J Biomed Health Inform ; 26(6): 2627-2636, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35085099

RESUMO

Localized disease heterogeneity on imaging extracted via radiomics approaches have recently been associated with disease prognosis and treatment response. Traditionally, radiomics analyses leverage texture operators to derive voxel- or region-wise feature values towards quantifying subtle variations in image appearance within a region-of-interest (ROI). With the goal of mining additional voxel-wise texture patterns from radiomic "expression maps", we introduce a new RADIomic Spatial TexturAl descripTor (RADISTAT). This was driven by the hypothesis that quantifying spatial organization of texture patterns within an ROI could allow for better capturing interactions between different tissue classes present in a given region; thus enabling more accurate characterization of disease or response phenotypes. RADISTAT involves: (a) robustly identifying sub-compartments of low, intermediate, and high radiomic expression (i.e. heterogeneity) in a feature map and (b) quantifying spatial organization of sub-compartments via graph interactions. RADISTAT was evaluated in two clinically challenging problems: (1) discriminating nodal/distant metastasis from metastasis-free rectal cancer patients on post-chemoradiation T2w MRI, and (2) distinguishing tumor progression from pseudo-progression in glioblastoma multiforme using post-chemoradiation T1w MRI. Across over 800 experiments, RADISTAT yielded a consistent discriminatory signature for tumor progression (GBM) and disease metastasis (RCa); where its sub-compartments were associated with pathologic tissue types (fibrosis or tumor, determined via fusion of MRI and pathology). In a multi-institutional setting for both clinical problems, RADISTAT resulted in higher classifier performance (11% improvement in AUC, on average) compared to radiomic descriptors. Furthermore, combining RADISTAT with radiomic descriptors resulted in significantly improved performance compared to using radiomic descriptors alone.


Assuntos
Glioblastoma , Humanos , Imageamento por Ressonância Magnética/métodos , Prognóstico
10.
Acad Radiol ; 29 Suppl 1: S223-S228, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-33160860

RESUMO

RATIONALE AND OBJECTIVES: Peritumoral features have been suggested to be useful in improving the prediction performance of radiomic models. The aim of this study is to systematically investigate the prediction performance improvement for sentinel lymph node (SLN) status in breast cancer from peritumoral features in radiomic analysis by exploring the effect of peritumoral region sizes. MATERIALS AND METHODS: This retrospective study was performed using dynamic contrast-enhanced MRI scans of 162 breast cancer patients. The effect of peritumoral features was evaluated in a radiomics pipeline for predicting SLN metastasis in breast cancer. Peritumoral regions were generated by dilating the tumor regions-of-interest (ROIs) manually annotated by two expert radiologists, with thicknesses of 2 mm, 4 mm, 6 mm, and 8 mm. The prediction models were established in the training set (∼67% of cases) using the radiomics pipeline with and without peritumoral features derived from different peritumoral thicknesses. The prediction performance was tested in an independent validation set (the remaining ∼33%). RESULTS: For this specific application, the accuracy in the validation set when using the two radiologists' ROIs could be both improved from 0.704 to 0.796 by incorporating peritumoral features. The choice of the peritumoral size could affect the level of improvement. CONCLUSION: This study systematically investigates the effect of peritumoral region sizes in radiomic analysis for prediction performance improvement. The choice of the peritumoral size is dependent on the ROI drawing and would affect the final prediction performance of radiomic models, suggesting that peritumoral features should be optimized in future radiomics studies.


Assuntos
Neoplasias da Mama , Linfonodo Sentinela , Mama/patologia , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/patologia , Feminino , Humanos , Metástase Linfática/diagnóstico por imagem , Estudos Retrospectivos , Linfonodo Sentinela/diagnóstico por imagem , Linfonodo Sentinela/patologia
11.
Annu Int Conf IEEE Eng Med Biol Soc ; 2021: 3998-4001, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34892107

RESUMO

Intratumor heterogeneity in glioblastoma (GBM) has been linked to adverse clinical outcomes including poor survival and sub-optimal response to therapies. Different techniques, such as radiomics, have been used to characterize GBM phenotype. However, the spatial diversity and the interaction between different sub-regions within the tumor (habitats) and its microenvironment has been relatively unexplored. Besides, existing approaches have mainly focused on the radiomic analysis within globally defined regions without considering local heterogeneity. In this paper, we developed a 3D spatial co-localization descriptor based on the adjacency of "habitats" to quantify the diversity of physiologically similar sub-regions on multi-protocol magnetic resonance imaging. We demonstrated the utility of this spatial phenotype descriptor in predicting overall patient survival. Our experimental results on N=236 treatment-naïve MRI scans suggest that the co-localization features in conjunction with traditional clinical measures, such as age and tumor volume, outperform texture based radiomic features. The presented descriptor provides a tool for more complete characterization of intratumor heterogeneity in solid cancers.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Biomarcadores , Neoplasias Encefálicas/diagnóstico por imagem , Ecossistema , Glioblastoma/diagnóstico por imagem , Humanos , Prognóstico , Microambiente Tumoral
12.
Cancers (Basel) ; 13(11)2021 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-34205005

RESUMO

The aim of this study is to evaluate whether NIS radiomics can distinguish lung adenocarcinomas from granulomas on non-contrast CT scans, and also to improve the performance of Lung-RADS by reclassifying benign nodules that were initially assessed as suspicious. The screening or standard diagnostic non-contrast CT scans of 362 patients was divided into training (St, N = 145), validation (Sv, N = 145), and independent validation (Siv, N = 62) sets from different institutions. Nodules were identified and manually segmented on CT images by a radiologist. A series of 264 features relating to the edge sharpness transition from the inside to the outside of the nodule were extracted. The top 10 features were used to train a linear discriminant analysis (LDA) machine learning classifier on St. In conjunction with the LDA classifier, NIS radiomics classified nodules with an AUC of 0.82 ± 0.04, 0.77, and 0.71 respectively on St, Sv, and Siv. We evaluated the ability of the NIS classifier to determine the proportion of the patients in Sv that were identified initially as suspicious by Lung-RADS but were reclassified as benign by applying the NIS scores. The NIS classifier was able to correctly reclassify 46% of those lesions that were actually benign but deemed suspicious by Lung-RADS alone on Sv.

13.
Br J Cancer ; 125(5): 641-657, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33958734

RESUMO

The natural history and treatment landscape of primary brain tumours are complicated by the varied tumour behaviour of primary or secondary gliomas (high-grade transformation of low-grade lesions), as well as the dilemmas with identification of radiation necrosis, tumour progression, and pseudoprogression on MRI. Radiomics and radiogenomics promise to offer precise diagnosis, predict prognosis, and assess tumour response to modern chemotherapy/immunotherapy and radiation therapy. This is achieved by a triumvirate of morphological, textural, and functional signatures, derived from a high-throughput extraction of quantitative voxel-level MR image metrics. However, the lack of standardisation of acquisition parameters and inconsistent methodology between working groups have made validations unreliable, hence multi-centre studies involving heterogenous study populations are warranted. We elucidate novel radiomic and radiogenomic workflow concepts and state-of-the-art descriptors in sub-visual MR image processing, with relevant literature on applications of such machine learning techniques in glioma management.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Genômica/métodos , Glioma/diagnóstico por imagem , Interpretação de Imagem Radiográfica Assistida por Computador/métodos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Glioma/genética , Glioma/patologia , Humanos , Aprendizado de Máquina , Imageamento por Ressonância Magnética , Gradação de Tumores , Prognóstico
14.
Med Image Anal ; 68: 101903, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33352373

RESUMO

Local spatial arrangement of nuclei in histopathology images of different cancer subtypes has been shown to have prognostic value. In order to capture localized nuclear architectural information, local cell cluster graph-based measurements have been proposed. However, conventional ways of cell graph construction only utilize nuclear spatial proximity, and do not differentiate between different cell types while constructing the graph. In this paper, we present feature-driven local cell cluster graph (FLocK), a new approach to constructing local cell graphs by simultaneously considering spatial proximity and attributes of the individual nuclei (e.g. shape, size, texture). In addition, we have designed a new set of quantitative graph-derived metrics to be extracted from FLocKs, in turn capturing the interplay between different proximally located clusters of nuclei. We have evaluated the efficacy of FLocK features extracted from H&E stained tissue images in two clinical applications: to classify short-term vs. long-term survival among patients of early stage non-small cell lung cancer (ES-NSCLC), and also to predict human papillomavirus (HPV) status of oropharyngeal squamous cell carcinoma (OP-SCCs). In the classification of long-term vs. short-term survival among patients of ES-NSCLC (training cohort, n = 434), the top 10 discriminative FLocK features related to the variation of FLocK size and intersected FLocK distance were identified, via Minimum Redundancy and Maximum Relevance (MRMR) selection, in 100 runs of 10-fold cross-validation, and in conjunction with a linear discriminant classifier yielded a mean AUC of 0.68 for predicting survival in the training cohort. This is better than other state-of-art histomorphometric and deep learning classifiers (cell cluster graphs (AUC = 0.62), global cell graph (AUC = 0.56), nuclear shape (AUC = 0.54), nuclear orientation (AUC = 0.61), AlexNet (AUC = 0.55), ResNet (AUC = 0.56)). The FLocK-based classifier yielded an AUC of 0.70 in an independent testing cohort (n = 150). The patients identified as "high-risk" had significantly poorer overall survival in the testing cohort, with a hazard ratio (95% confidence interval) of 2.24 (1.24-4.05), p = 0.01144). In the classification of HPV status of OP-SCC, the top three FLocK features pertaining to the portion of intersected FLocKs were used to construct a classifier, which yielded an AUC of 0.80 in the training cohort (n = 50), and an accuracy of 0.78 in an independent testing cohort (n = 35). The combination of FLocK measurements with cell cluster graphs, nuclear orientation, and nuclear shape improved the training AUC to 0.87, 0.91 and 0.85, respectively. Deep learning approaches yielded marginally better performance than the FLocK-based classifier in this application, with AUC = 0.78 for AlexNet, AUC = 0.81 for ResNet, and AUC = 0.76 for FLocK-based classifier in the testing cohort. However, the combination of two hand-crafted features: FLocK and nuclear orientation yielded a better performance (AUC = 0.84). FLocK provides a unique and quantitative way to analyze histology images of solid tumors and interrogate tumor morphology from a different aspect than existing histomorphometrics. The source code can be accessed at https://github.com/hacylu/FLocK.


Assuntos
Alphapapillomavirus , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Neoplasias Orofaríngeas , Infecções por Papillomavirus , Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Humanos , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Orofaríngeas/diagnóstico por imagem , Papillomaviridae , Infecções por Papillomavirus/diagnóstico por imagem , Prognóstico
15.
Neuro Oncol ; 23(2): 251-263, 2021 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-33068415

RESUMO

BACKGROUND: Recent epidemiological studies have suggested that sexual dimorphism influences treatment response and prognostic outcome in glioblastoma (GBM). To this end, we sought to (i) identify distinct sex-specific radiomic phenotypes-from tumor subcompartments (peritumoral edema, enhancing tumor, and necrotic core) using pretreatment MRI scans-that are prognostic of overall survival (OS) in GBMs, and (ii) investigate radiogenomic associations of the MRI-based phenotypes with corresponding transcriptomic data, to identify the signaling pathways that drive sex-specific tumor biology and treatment response in GBM. METHODS: In a retrospective setting, 313 GBM patients (male = 196, female = 117) were curated from multiple institutions for radiomic analysis, where 130 were used for training and independently validated on a cohort of 183 patients. For the radiogenomic analysis, 147 GBM patients (male = 94, female = 53) were used, with 125 patients in training and 22 cases for independent validation. RESULTS: Cox regression models of radiomic features from gadolinium T1-weighted MRI allowed for developing more precise prognostic models, when trained separately on male and female cohorts. Our radiogenomic analysis revealed higher expression of Laws energy features that capture spots and ripple-like patterns (representative of increased heterogeneity) from the enhancing tumor region, as well as aggressive biological processes of cell adhesion and angiogenesis to be more enriched in the "high-risk" group of poor OS in the male population. In contrast, higher expressions of Laws energy features (which detect levels and edges) from the necrotic core with significant involvement of immune related signaling pathways was observed in the "low-risk" group of the female population. CONCLUSIONS: Sexually dimorphic radiogenomic models could help risk-stratify GBM patients for personalized treatment decisions.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/genética , Feminino , Glioblastoma/diagnóstico por imagem , Glioblastoma/genética , Humanos , Imageamento por Ressonância Magnética , Masculino , Prognóstico , Estudos Retrospectivos
16.
IEEE Trans Biomed Eng ; 68(6): 1777-1786, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32822291

RESUMO

Diabetic Macular Edema (DME) and macular edema secondary to retinal occlusion (RVO) are the two most common retinal vascular causes of visual impairment and leading cause of worldwide vision loss. The blood-retinal barrier is the key barrier for maintaining fluid balance within the retinal tissue. Vascular Endothelial Growth Factor (VEGF) has a significant role in the permeability of the blood-retinal barrier, which also leads to appearance of leakage foci. Intravitreal anti-VEGF therapy is the current gold standard treatment and has been demonstrated to improve macular thickening, improve vision acuity and reduce vascular leakage. However, treatment response and required dosing interval can vary widely across patients. Given the role of the blood-retinal barrier and vascular leakage in the pathogenesis of these disorders, the goal of this study was to present and evaluate new computer extracted features relating to morphology, spatial architecture and tortuosity of vessels and leakages from baseline ultra-widefield fluorescein angiography (UWFA) images. Specifically, we sought to evaluate the role of these computer extracted features from baseline UWFA images. Notably, these UWFA images were obtained from IRB-approved PERMEATE clinical trial [1], [2] to distinguish eyes tolerating extended dosing intervals (n = 16) who are referred to as non-rebounders and those who require more frequent dosing (n = 12) and are called rebounders based on visual acuity loss with extended dosing challenges. A total of 64 features encapsulating different morphological and geometrical attributes of leakage patches including the anatomical (shape, size, density, area, minor and major axis, orientation, area, extent ratio, perimeter, radii) and geometrical characteristics (the proximity of each leakage foci to main vessels, to other leakage foci and to optical disc) as well as 54 tortuosity features (tortuosity of whole vessel network, local tortuosity of vessels in the vicinity of leakage foci) were extracted. The most significant and predictive biomarkers related to treatment response were proximity of leakage nodes to major and minor eye vessels as well as local vasculature tortuosity in the vicinity of the leakages. The imaging features were then used in conjunction with a Linear Discriminant Analysis (LDA) classifier to distinguish rebounders from non-rebounders. The 3-fold cross-validated Area Under Curve (AUC) was found to be 0.82 for the morphological based features and 0.85 for the tortuosity based features. Our findings suggest higher variation in leakage node proximity to retinal vessels in eyes tolerating extended interval dosing. In contrast, eyes with increased local vascular tortuosity demonstrated less tolerance of increased dosing interval. Moreover, a class activation map generated by a deep learning model identified regions that corresponded to regions of leakages proximal to the vessels, providing confirmation of the validity of predictive image features extracted from these regions in this study.


Assuntos
Retinopatia Diabética , Edema Macular , Inibidores da Angiogênese , Retinopatia Diabética/diagnóstico por imagem , Retinopatia Diabética/tratamento farmacológico , Angiofluoresceinografia , Humanos , Edema Macular/diagnóstico por imagem , Edema Macular/tratamento farmacológico , Receptores de Fatores de Crescimento do Endotélio Vascular , Proteínas Recombinantes de Fusão , Vasos Retinianos/diagnóstico por imagem , Tomografia de Coerência Óptica , Fator A de Crescimento do Endotélio Vascular
17.
Eur Radiol ; 31(1): 379-391, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32700021

RESUMO

OBJECTIVES: To evaluate short-term test-retest repeatability of a deep learning architecture (U-Net) in slice- and lesion-level detection and segmentation of clinically significant prostate cancer (csPCa: Gleason grade group > 1) using diffusion-weighted imaging fitted with monoexponential function, ADCm. METHODS: One hundred twelve patients with prostate cancer (PCa) underwent 2 prostate MRI examinations on the same day. PCa areas were annotated using whole mount prostatectomy sections. Two U-Net-based convolutional neural networks were trained on three different ADCm b value settings for (a) slice- and (b) lesion-level detection and (c) segmentation of csPCa. Short-term test-retest repeatability was estimated using intra-class correlation coefficient (ICC(3,1)), proportionate agreement, and dice similarity coefficient (DSC). A 3-fold cross-validation was performed on training set (N = 78 patients) and evaluated for performance and repeatability on testing data (N = 34 patients). RESULTS: For the three ADCm b value settings, repeatability of mean ADCm of csPCa lesions was ICC(3,1) = 0.86-0.98. Two CNNs with U-Net-based architecture demonstrated ICC(3,1) in the range of 0.80-0.83, agreement of 66-72%, and DSC of 0.68-0.72 for slice- and lesion-level detection and segmentation of csPCa. Bland-Altman plots suggest that there is no systematic bias in agreement between inter-scan ground truth segmentation repeatability and segmentation repeatability of the networks. CONCLUSIONS: For the three ADCm b value settings, two CNNs with U-Net-based architecture were repeatable for the problem of detection of csPCa at the slice-level. The network repeatability in segmenting csPCa lesions is affected by inter-scan variability and ground truth segmentation repeatability and may thus improve with better inter-scan reproducibility. KEY POINTS: • For the three ADCm b value settings, two CNNs with U-Net-based architecture were repeatable for the problem of detection of csPCa at the slice-level. • The network repeatability in segmenting csPCa lesions is affected by inter-scan variability and ground truth segmentation repeatability and may thus improve with better inter-scan reproducibility.


Assuntos
Aprendizado Profundo , Neoplasias da Próstata , Imagem de Difusão por Ressonância Magnética , Humanos , Masculino , Redes Neurais de Computação , Neoplasias da Próstata/diagnóstico por imagem , Reprodutibilidade dos Testes
18.
Br J Ophthalmol ; 105(8): 1155-1160, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-32816791

RESUMO

AIM: To evaluate the potential of radiomics-based ultra-widefield fluorescein angiography (UWFA)-derived imaging biomarkers in retinal vascular disease for predicting therapeutic durability of intravitreal aflibercept injection (IAI). METHODS: The Peripheral and Macular Retinal Vascular Perfusion and Leakage Dynamics in Diabetic Macular Edema and Retinal Venous Occlusions During Intravitreal Aflibercept Injection (IAI) Treatment for Retinal Edema (PERMEATE) study prospectively evaluated quantitative UWFA dynamics in diabetic macular oedema or macular oedema secondary to retinal vascular occlusion. 27 treatment-naïve eyes were treated with 2 mg IAI q4 weeks for the first 6 months, and then administered q8 weeks. Morphological and graph-based attributes were used to model the spatial distribution of leakage areas, while tortuosity measures were used to model the vessel network disorder. Eyes were grouped based on functional tolerance of the first 8-week treatment interval challenge. 'Non-rebounders' (N=15) maintained/improved best-corrected visual acuity (BCVA) following the 8-week challenge. 'Rebounders' (N=12) exhibited worsened BVCA. The image biomarkers were used with a machine learning classifier to preliminarily evaluate their ability to predict BCVA stability. RESULTS: Two new UWFA image-derived biomarkers were identified and extracted. The cross-validated area under the receiver operating characteristic curve (AUC) was 0.77±0.14 using baseline leakage distribution features and 0.73±0.10 for the UWFA baseline tortuosity measures. Additionally, the change in vascular tortuosity between month 4 and baseline yielded an AUC of 0.73±0.08. Three baseline clinical features of letter score, macular volume and central subfield thickness yielded a corresponding AUC of 0.42±0.09. CONCLUSIONS: Two computer-extracted UWFA radiomics-based descriptors were identified as potential biomarkers for predicting treatment durability and tolerance of longer treatment intervals. Conventional treatment parameters were not significantly different between these same groups.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Permeabilidade Capilar/fisiologia , Retinopatia Diabética/tratamento farmacológico , Angiofluoresceinografia , Edema Macular/tratamento farmacológico , Receptores de Fatores de Crescimento do Endotélio Vascular/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Vasos Retinianos/patologia , Idoso , Área Sob a Curva , Biomarcadores , Barreira Hematorretiniana/fisiologia , Retinopatia Diabética/diagnóstico , Retinopatia Diabética/fisiopatologia , Feminino , Humanos , Injeções Intravítreas , Edema Macular/diagnóstico , Edema Macular/fisiopatologia , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Curva ROC , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Acuidade Visual/fisiologia
19.
Lancet Digit Health ; 2(3): e116-e128, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-33334576

RESUMO

BACKGROUND: Use of adjuvant chemotherapy in patients with early-stage lung cancer is controversial because no definite biomarker exists to identify patients who would receive added benefit from it. We aimed to develop and validate a quantitative radiomic risk score (QuRiS) and associated nomogram (QuRNom) for early-stage non-small cell lung cancer (NSCLC) that is prognostic of disease-free survival and predictive of the added benefit of adjuvant chemotherapy following surgery. METHODS: We did a retrospective multicohort study of individuals with early-stage NSCLC (stage I and II) who either received surgery alone or surgery plus adjuvant chemotherapy. We selected patients for whom we had available pre-treatment diagnostic CT scans and corresponding survival information. We used radiomic texture features derived from within and outside the primary lung nodule on chest CT scans of patients from the Cleveland Clinic Foundation (Cleveland, OH, USA; cohort D1) to develop QuRiS. A least absolute shrinkage and selection operator-Cox regularisation model was used for data dimension reduction, feature selection, and QuRiS construction. QuRiS was independently validated on a cohort of patients from the University of Pennsylvania (Philadephia, PA, USA; cohort D2) and a cohort of patients whose CT scans were derived from The Cancer Imaging Archive (cohort D3). QuRNom was constructed by integrating QuRiS with tumour and node descriptors (according to the tumour, node, metastasis staging system) and lymphovascular invasion. The primary endpoint of the study was the assessment of the performance of QuRiS and QuRNom in predicting disease-free survival. The added benefit of adjuvant chemotherapy estimated using QuRiS and QuRNom was validated by comparing patients who received adjuvant chemotherapy versus patients who underwent surgery alone in cohorts D1-D3. FINDINGS: We included: 329 patients in cohort D1 (73 [22%] had surgery plus adjuvant chemotherapy and 256 (78%) had surgery alone); 114 patients in cohort D2 (33 [29%] had surgery plus adjuvant chemotherapy and 81 (71%) had surgery alone); and 82 patients in cohort D3 (24 [29%] had surgery plus adjuvant chemotherapy and 58 (71%) had surgery alone). QuRiS comprised three intratumoral and 10 peritumoral CT-radiomic features and was found to be significantly associated with disease-free survival (ie, prognostic validation of QuRiS; hazard ratio for predicted high-risk vs predicted low-risk groups 1·56, 95% CI 1·08-2·23, p=0·016 for cohort D1; 2·66, 1·24-5·68, p=0·011 for cohort D2; and 2·67, 1·39-5·11, p=0·0029 for cohort D3). To validate the predictive performance of QuRiS, patients were partitioned into three risk groups (high, intermediate, and low risk) on the basis of their corresponding QuRiS. Patients in the high-risk group were observed to have significantly longer survival with adjuvant chemotherapy than patients who underwent surgery alone (0·27, 0·08-0·95, p=0·042, for cohort D1; 0·08, 0·01-0·42, p=0·0029, for cohorts D2 and D3 combined). As concerns QuRNom, the nomogram-estimated survival benefit was predictive of the actual efficacy of adjuvant chemotherapy (0·25, 0·12-0·55, p<0·0001, for cohort D1; 0·13, <0·01-0·99, p=0·0019 for cohort D3). INTERPRETATION: QuRiS and QuRNom were validated as being prognostic of disease-free survival and predictive of the added benefit of adjuvant chemotherapy, especially in clinically defined low-risk groups. Since QuRiS is based on routine chest CT imaging, with additional multisite independent validation it could potentially be employed for decision management in non-invasive treatment of resectable lung cancer. FUNDING: National Cancer Institute of the US National Institutes of Health, National Center for Research Resources, US Department of Veterans Affairs Biomedical Laboratory Research and Development Service, Department of Defence, National Institute of Diabetes and Digestive and Kidney Diseases, Wallace H Coulter Foundation, Case Western Reserve University, and Dana Foundation.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/tratamento farmacológico , Tomografia Computadorizada por Raios X , Idoso , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/cirurgia , Quimioterapia Adjuvante , Estudos de Coortes , Feminino , Humanos , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/cirurgia , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Valor Preditivo dos Testes , Prognóstico , Estudos Retrospectivos
20.
Front Comput Neurosci ; 14: 563439, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33381018

RESUMO

A significant challenge in Glioblastoma (GBM) management is identifying pseudo-progression (PsP), a benign radiation-induced effect, from tumor recurrence, on routine imaging following conventional treatment. Previous studies have linked tumor lobar presence and laterality to GBM outcomes, suggesting that disease etiology and progression in GBM may be impacted by tumor location. Hence, in this feasibility study, we seek to investigate the following question: Can tumor location on treatment-naïve MRI provide early cues regarding likelihood of a patient developing pseudo-progression vs. tumor recurrence? In this study, 74 pre-treatment Glioblastoma MRI scans with PsP (33) and tumor recurrence (41) were analyzed. First, enhancing lesion on Gd-T1w MRI and peri-lesional hyperintensities on T2w/FLAIR were segmented by experts and then registered to a brain atlas. Using patients from the two phenotypes, we construct two atlases by quantifying frequency of occurrence of enhancing lesion and peri-lesion hyperintensities, by averaging voxel intensities across the population. Analysis of differential involvement was then performed to compute voxel-wise significant differences (p-value < 0.05) across the atlases. Statistically significant clusters were finally mapped to a structural atlas to provide anatomic localization of their location. Our results demonstrate that patients with tumor recurrence showed prominence of their initial tumor in the parietal lobe, while patients with PsP showed a multi-focal distribution of the initial tumor in the frontal and temporal lobes, insula, and putamen. These preliminary results suggest that lateralization of pre-treatment lesions toward certain anatomical areas of the brain may allow to provide early cues regarding assessing likelihood of occurrence of pseudo-progression from tumor recurrence on MRI scans.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA