Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 152
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 173(2): 338-354.e15, 2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29625051

RESUMEN

Cancer progression involves the gradual loss of a differentiated phenotype and acquisition of progenitor and stem-cell-like features. Here, we provide novel stemness indices for assessing the degree of oncogenic dedifferentiation. We used an innovative one-class logistic regression (OCLR) machine-learning algorithm to extract transcriptomic and epigenetic feature sets derived from non-transformed pluripotent stem cells and their differentiated progeny. Using OCLR, we were able to identify previously undiscovered biological mechanisms associated with the dedifferentiated oncogenic state. Analyses of the tumor microenvironment revealed unanticipated correlation of cancer stemness with immune checkpoint expression and infiltrating immune cells. We found that the dedifferentiated oncogenic phenotype was generally most prominent in metastatic tumors. Application of our stemness indices to single-cell data revealed patterns of intra-tumor molecular heterogeneity. Finally, the indices allowed for the identification of novel targets and possible targeted therapies aimed at tumor differentiation.


Asunto(s)
Desdiferenciación Celular/genética , Aprendizaje Automático , Neoplasias/patología , Carcinogénesis , Metilación de ADN , Bases de Datos Genéticas , Epigénesis Genética , Humanos , MicroARNs/metabolismo , Metástasis de la Neoplasia , Neoplasias/genética , Células Madre/citología , Células Madre/metabolismo , Transcriptoma , Microambiente Tumoral
2.
Bioinformatics ; 40(1)2024 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-38134424

RESUMEN

MOTIVATION: Drug-target interaction (DTI) prediction is a relevant but challenging task in the drug repurposing field. In-silico approaches have drawn particular attention as they can reduce associated costs and time commitment of traditional methodologies. Yet, current state-of-the-art methods present several limitations: existing DTI prediction approaches are computationally expensive, thereby hindering the ability to use large networks and exploit available datasets and, the generalization to unseen datasets of DTI prediction methods remains unexplored, which could potentially improve the development processes of DTI inferring approaches in terms of accuracy and robustness. RESULTS: In this work, we introduce GeNNius (Graph Embedding Neural Network Interaction Uncovering System), a Graph Neural Network (GNN)-based method that outperforms state-of-the-art models in terms of both accuracy and time efficiency across a variety of datasets. We also demonstrated its prediction power to uncover new interactions by evaluating not previously known DTIs for each dataset. We further assessed the generalization capability of GeNNius by training and testing it on different datasets, showing that this framework can potentially improve the DTI prediction task by training on large datasets and testing on smaller ones. Finally, we investigated qualitatively the embeddings generated by GeNNius, revealing that the GNN encoder maintains biological information after the graph convolutions while diffusing this information through nodes, eventually distinguishing protein families in the node embedding space. AVAILABILITY AND IMPLEMENTATION: GeNNius code is available at https://github.com/ubioinformat/GeNNius.


Asunto(s)
Sistemas de Liberación de Medicamentos , Reposicionamiento de Medicamentos , Interacciones Farmacológicas , Difusión , Redes Neurales de la Computación
3.
Hum Mol Genet ; 31(13): 2164-2184, 2022 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-35094088

RESUMEN

Sotos syndrome (SS), the most common overgrowth with intellectual disability (OGID) disorder, is caused by inactivating germline mutations of NSD1, which encodes a histone H3 lysine 36 methyltransferase. To understand how NSD1 inactivation deregulates transcription and DNA methylation (DNAm), and to explore how these abnormalities affect human development, we profiled transcription and DNAm in SS patients and healthy control individuals. We identified a transcriptional signature that distinguishes individuals with SS from controls and was also deregulated in NSD1-mutated cancers. Most abnormally expressed genes displayed reduced expression in SS; these downregulated genes consisted mostly of bivalent genes and were enriched for regulators of development and neural synapse function. DNA hypomethylation was strongly enriched within promoters of transcriptionally deregulated genes: overexpressed genes displayed hypomethylation at their transcription start sites while underexpressed genes featured hypomethylation at polycomb binding sites within their promoter CpG island shores. SS patients featured accelerated molecular aging at the levels of both transcription and DNAm. Overall, these findings indicate that NSD1-deposited H3K36 methylation regulates transcription by directing promoter DNA methylation, partially by repressing polycomb repressive complex 2 (PRC2) activity. These findings could explain the phenotypic similarity of SS to OGID disorders that are caused by mutations in PRC2 complex-encoding genes.


Asunto(s)
Síndrome de Sotos , Metilación de ADN/genética , Genes del Desarrollo , Histona Metiltransferasas/genética , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Mutación , Síndrome de Sotos/genética
4.
Eur Radiol ; 32(3): 2030-2040, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34564745

RESUMEN

OBJECTIVES: To assess the precision of MRI radiomics features in hepatocellular carcinoma (HCC) tumors and liver parenchyma. METHODS: The study population consisted of 55 patients, including 16 with untreated HCCs, who underwent two repeat contrast-enhanced abdominal MRI exams within 1 month to evaluate: (1) test-retest repeatability using the same MRI system (n = 28, 10 HCCs); (2) inter-platform reproducibility between different MRI systems (n = 27, 6 HCCs); (3) inter-observer reproducibility (n = 16, 16 HCCs). Shape and 1st- and 2nd-order radiomics features were quantified on pre-contrast T1-weighted imaging (WI), T1WI portal venous phase (pvp), T2WI, and ADC (apparent diffusion coefficient), on liver regions of interest (ROIs) and HCC volumes of interest (VOIs). Precision was assessed by calculating intraclass correlation coefficient (ICC), concordance correlation coefficient (CCC), and coefficient of variation (CV). RESULTS: There was moderate to excellent test-retest repeatability of shape and 1st- and 2nd-order features for all sequences in HCCs (ICC: 0.53-0.99; CV: 3-29%), and moderate to good test-retest repeatability of 1st- and 2nd-order features for T1WI sequences, and 2nd-order features for T2WI in the liver (ICC: 0.53-0.73; CV: 12-19%). There was poor inter-platform reproducibility for all features and sequences, except for shape and 1st-order features on T1WI in HCCs (CCC: 0.58-0.99; CV: 3-15%). Good to excellent inter-observer reproducibility was found for all features and sequences in HCCs (CCC: 0.80-0.99; CV: 4-15%) and moderate to good for liver (CCC: 0.45-0.86; CV: 6-25%). CONCLUSIONS: MRI radiomics features have acceptable repeatability in the liver and HCC when using the same MRI system and across readers but have low reproducibility across MR systems, except for shape and 1st-order features on T1WI. Data must be interpreted with caution when performing multiplatform radiomics studies. KEY POINTS: • MRI radiomics features have acceptable repeatability when using the same MRI system but less reproducible when using different MRI platforms. • MRI radiomics features extracted from T1 weighted-imaging show greater stability across exams than T2 weighted-imaging and ADC. • Inter-observer reproducibility of MRI radiomics features was found to be good in HCC tumors and acceptable in liver parenchyma.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Carcinoma Hepatocelular/diagnóstico por imagen , Humanos , Neoplasias Hepáticas/diagnóstico por imagen , Imagen por Resonancia Magnética , Reproducibilidad de los Resultados , Estudios Retrospectivos
5.
Br J Cancer ; 125(3): 309-310, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33782563

RESUMEN

Meta-learning is showing promise in recent genomic studies in oncology. Meta-learning can facilitate transfer learning and reduce the amount of data that is needed in a target domain by transferring knowledge from abundant genomic data in different source domains enabling the use of AI in data scarce scenarios.


Asunto(s)
Biología Computacional/métodos , Genómica , Neoplasias/genética , Inteligencia Artificial , Humanos , Metadatos
6.
Bioinformatics ; 36(2): 558-567, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31287491

RESUMEN

MOTIVATION: Gene regulatory networks describe the regulatory relationships among genes, and developing methods for reverse engineering these networks is an ongoing challenge in computational biology. The majority of the initially proposed methods for gene regulatory network discovery create a network of genes and then mine it in order to uncover previously unknown regulatory processes. More recent approaches have focused on inferring modules of co-regulated genes, linking these modules with regulatory genes and then mining them to discover new molecular biology. RESULTS: In this work we analyze module-based network approaches to build gene regulatory networks, and compare their performance to single gene network approaches. In the process, we propose a novel approach to estimate gene regulatory networks drawing from the module-based methods. We show that generating modules of co-expressed genes which are predicted by a sparse set of regulators using a variational Bayes method, and then building a bipartite graph on the generated modules using sparse regression, yields more informative networks than previous single and module-based network approaches as measured by: (i) the rate of enriched gene sets, (ii) a network topology assessment, (iii) ChIP-Seq evidence and (iv) the KnowEnG Knowledge Network collection of previously characterized gene-gene interactions. AVAILABILITY AND IMPLEMENTATION: The code is written in R and can be downloaded from https://github.com/mikelhernaez/linker. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Algoritmos , Redes Reguladoras de Genes , Teorema de Bayes , Biología Computacional , Perfilación de la Expresión Génica
7.
BMC Cancer ; 21(1): 1246, 2021 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-34798858

RESUMEN

BACKGROUND: Atypical tumor response patterns during immune checkpoint inhibitor therapy pose a challenge to clinicians and investigators in immuno-oncology practice. This study evaluated tumor burden dynamics to identify imaging biomarkers for treatment response and overall survival (OS) in advanced gastrointestinal malignancies treated with PD-1/PD-L1 inhibitors. METHODS: This retrospective study enrolled a total of 198 target lesions in 75 patients with advanced gastrointestinal malignancies treated with PD-1/PD-L1 inhibitors between January 2017 and March 2021. Tumor diameter changes as defined by immunotherapy Response Evaluation Criteria in Solid Tumors (iRECIST) were studied to determine treatment response and association with OS. RESULTS: Based on the best overall response, the tumor diameter ranged from - 100 to + 135.3% (median: - 9.6%). The overall response rate was 32.0% (24/75), and the rate of durable disease control for at least 6 months was 30.7% (23/75, one (iCR, immune complete response) or 20 iPR (immune partial response), or 2iSD (immune stable disease). Using univariate analysis, patients with a tumor diameter maintaining a < 20% increase (48/75, 64.0%) from baseline had longer OS than those with ≥20% increase (27/75, 36.0%) and, a reduced risk of death (median OS: 80 months vs. 48 months, HR = 0.22, P = 0.034). The differences in age (HR = 1.09, P = 0.01), combined surgery (HR = 0.15, P = 0.01) and cancer type (HR = 0.23, P = 0.001) were significant. In multivariable analysis, patients with a tumor diameter with a < 20% increase had notably reduced hazards of death (HR = 0.15, P = 0.01) after adjusting for age, combined surgery, KRAS status, cancer type, mismatch repair (MMR) status, treatment course and cancer differentiation. Two patients (2.7%) showed pseudoprogression. CONCLUSIONS: Tumor diameter with a < 20% increase from baseline during therapy in gastrointestinal malignancies was associated with therapeutic benefit and longer OS and may serve as a practical imaging marker for treatment response, clinical outcome and treatment decision making.


Asunto(s)
Neoplasias Gastrointestinales , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Criterios de Evaluación de Respuesta en Tumores Sólidos , Carga Tumoral , Adulto , Factores de Edad , Análisis de Varianza , Reparación de la Incompatibilidad de ADN , Femenino , Neoplasias Gastrointestinales/tratamiento farmacológico , Neoplasias Gastrointestinales/inmunología , Neoplasias Gastrointestinales/mortalidad , Neoplasias Gastrointestinales/patología , Genes ras , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos
8.
Gastroenterology ; 157(2): 537-551.e9, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30978357

RESUMEN

BACKGROUND & AIMS: The mechanisms of hepatitis C virus (HCV) infection, liver disease progression, and hepatocarcinogenesis are only partially understood. We performed genomic, proteomic, and metabolomic analyses of HCV-infected cells and chimeric mice to learn more about these processes. METHODS: Huh7.5.1dif (hepatocyte-like cells) were infected with culture-derived HCV and used in RNA sequencing, proteomic, metabolomic, and integrative genomic analyses. uPA/SCID (urokinase-type plasminogen activator/severe combined immunodeficiency) mice were injected with serum from HCV-infected patients; 8 weeks later, liver tissues were collected and analyzed by RNA sequencing and proteomics. Using differential expression, gene set enrichment analyses, and protein interaction mapping, we identified pathways that changed in response to HCV infection. We validated our findings in studies of liver tissues from 216 patients with HCV infection and early-stage cirrhosis and paired biopsy specimens from 99 patients with hepatocellular carcinoma, including 17 patients with histologic features of steatohepatitis. Cirrhotic liver tissues from patients with HCV infection were classified into 2 groups based on relative peroxisome function; outcomes assessed included Child-Pugh class, development of hepatocellular carcinoma, survival, and steatohepatitis. Hepatocellular carcinomas were classified according to steatohepatitis; the outcome was relative peroxisomal function. RESULTS: We quantified 21,950 messenger RNAs (mRNAs) and 8297 proteins in HCV-infected cells. Upon HCV infection of hepatocyte-like cells and chimeric mice, we observed significant changes in levels of mRNAs and proteins involved in metabolism and hepatocarcinogenesis. HCV infection of hepatocyte-like cells significantly increased levels of the mRNAs, but not proteins, that regulate the innate immune response; we believe this was due to the inhibition of translation in these cells. HCV infection of hepatocyte-like cells increased glucose consumption and metabolism and the STAT3 signaling pathway and reduced peroxisome function. Peroxisomes mediate ß-oxidation of very long-chain fatty acids; we found intracellular accumulation of very long-chain fatty acids in HCV-infected cells, which is also observed in patients with fatty liver disease. Cells in livers from HCV-infected mice had significant reductions in levels of the mRNAs and proteins associated with peroxisome function, indicating perturbation of peroxisomes. We found that defects in peroxisome function were associated with outcomes and features of HCV-associated cirrhosis, fatty liver disease, and hepatocellular carcinoma in patients. CONCLUSIONS: We performed combined transcriptome, proteome, and metabolome analyses of liver tissues from HCV-infected hepatocyte-like cells and HCV-infected mice. We found that HCV infection increases glucose metabolism and the STAT3 signaling pathway and thereby reduces peroxisome function; alterations in the expression levels of peroxisome genes were associated with outcomes of patients with liver diseases. These findings provide insights into liver disease pathogenesis and might be used to identify new therapeutic targets.


Asunto(s)
Hepacivirus/patogenicidad , Hepatitis C Crónica/patología , Hepatocitos/patología , Hígado/patología , Animales , Línea Celular Tumoral , Conjuntos de Datos como Asunto , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Glucosa/metabolismo , Hepatitis C Crónica/metabolismo , Hepatitis C Crónica/virología , Hepatocitos/trasplante , Hepatocitos/virología , Humanos , Hígado/citología , Hígado/virología , Metabolómica , Ratones , Peroxisomas/metabolismo , Peroxisomas/patología , Proteómica , Factor de Transcripción STAT3/metabolismo , Quimera por Trasplante
9.
Bioinformatics ; 35(14): i446-i454, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-31510656

RESUMEN

MOTIVATION: Estimating the future course of patients with cancer lesions is invaluable to physicians; however, current clinical methods fail to effectively use the vast amount of multimodal data that is available for cancer patients. To tackle this problem, we constructed a multimodal neural network-based model to predict the survival of patients for 20 different cancer types using clinical data, mRNA expression data, microRNA expression data and histopathology whole slide images (WSIs). We developed an unsupervised encoder to compress these four data modalities into a single feature vector for each patient, handling missing data through a resilient, multimodal dropout method. Encoding methods were tailored to each data type-using deep highway networks to extract features from clinical and genomic data, and convolutional neural networks to extract features from WSIs. RESULTS: We used pancancer data to train these feature encodings and predict single cancer and pancancer overall survival, achieving a C-index of 0.78 overall. This work shows that it is possible to build a pancancer model for prognosis that also predicts prognosis in single cancer sites. Furthermore, our model handles multiple data modalities, efficiently analyzes WSIs and represents patient multimodal data flexibly into an unsupervised, informative representation. We thus present a powerful automated tool to accurately determine prognosis, a key step towards personalized treatment for cancer patients. AVAILABILITY AND IMPLEMENTATION: https://github.com/gevaertlab/MultimodalPrognosis.


Asunto(s)
Aprendizaje Profundo , Neoplasias , Genoma , Humanos , Neoplasias/genética , Redes Neurales de la Computación
10.
PLoS Comput Biol ; 15(7): e1007245, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31356589

RESUMEN

Aberrant DNA methylation disrupts normal gene expression in cancer and broadly contributes to oncogenesis. We previously developed MethylMix, a model-based algorithmic approach to identify epigenetically regulated driver genes. MethylMix identifies genes where methylation likely executes a functional role by using transcriptomic data to select only methylation events that can be linked to changes in gene expression. However, given that proteins more closely link genotype to phenotype recent high-throughput proteomic data provides an opportunity to more accurately identify functionally relevant abnormal methylation events. Here we present a MethylMix analysis that refines nominations for epigenetic driver genes by leveraging quantitative high-throughput proteomic data to select only genes where DNA methylation is predictive of protein abundance. Applying our algorithm across three cancer cohorts we find that using protein abundance data narrows candidate nominations, where the effect of DNA methylation is often buffered at the protein level. Next, we find that MethylMix genes predictive of protein abundance are enriched for biological processes involved in cancer including functions involved in epithelial and mesenchymal transition. Moreover, our results are also enriched for tumor markers which are predictive of clinical features like tumor stage and we find clustering using MethylMix genes predictive of protein abundance captures cancer subtypes.


Asunto(s)
Metilación de ADN , Neoplasias/genética , Neoplasias/metabolismo , Proteoma/genética , Algoritmos , Biomarcadores de Tumor/genética , Biología Computacional , Progresión de la Enfermedad , Epigénesis Genética , Transición Epitelial-Mesenquimal/genética , Transición Epitelial-Mesenquimal/fisiología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Modelos Genéticos , Familia de Multigenes , Neoplasias/patología
11.
Chin J Cancer Res ; 32(1): 62-71, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32194306

RESUMEN

OBJECTIVE: To develop and validate a computed tomography (CT)-based radiomics nomogram for predicting human epidermal growth factor receptor 2 (HER2) status in patients with gastric cancer. METHODS: This retrospective study included 134 patients with gastric cancer (HER2-negative: n=87; HER2-positive: n=47) from April 2013 to March 2018, who were then randomly divided into training (n=94) and validation (n=40) cohorts. Radiomics features were obtained from the CT images showing gastric cancer. Least absolute shrinkage and selection operator (LASSO) regression analysis was utilized for building the radiomics signature. A multivariable logistic regression method was applied to develop a prediction model incorporating the radiomics signature and independent clinicopathologic risk predictors, which were then visualized as a radiomics nomogram. The predictive performance of the nomogram was assessed in the training and validation cohorts. RESULTS: The radiomics signature was significantly associated with HER2 status in both training (P<0.001) and validation (P=0.023) cohorts. The prediction model that incorporated the radiomics signature and carcinoembryonic antigen (CEA) level demonstrated good discriminative performance for HER2 status prediction, with an area under the curve (AUC) of 0.799 [95% confidence interval (95% CI): 0.704-0.894] in the training cohort and 0.771 (95% CI: 0.607-0.934) in the validation cohort. The calibration curve of the radiomics nomogram also showed good calibration. Decision curve analysis showed that the radiomics nomogram was useful. CONCLUSIONS: We built and validated a radiomics nomogram with good performance for HER2 status prediction in gastric cancer. This radiomics nomogram could serve as a non-invasive tool to predict HER2 status and guide clinical treatment.

12.
Eur Respir J ; 53(3)2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30635290

RESUMEN

Epidermal growth factor receptor (EGFR) genotyping is critical for treatment guidelines such as the use of tyrosine kinase inhibitors in lung adenocarcinoma. Conventional identification of EGFR genotype requires biopsy and sequence testing which is invasive and may suffer from the difficulty of accessing tissue samples. Here, we propose a deep learning model to predict EGFR mutation status in lung adenocarcinoma using non-invasive computed tomography (CT).We retrospectively collected data from 844 lung adenocarcinoma patients with pre-operative CT images, EGFR mutation and clinical information from two hospitals. An end-to-end deep learning model was proposed to predict the EGFR mutation status by CT scanning.By training in 14 926 CT images, the deep learning model achieved encouraging predictive performance in both the primary cohort (n=603; AUC 0.85, 95% CI 0.83-0.88) and the independent validation cohort (n=241; AUC 0.81, 95% CI 0.79-0.83), which showed significant improvement over previous studies using hand-crafted CT features or clinical characteristics (p<0.001). The deep learning score demonstrated significant differences in EGFR-mutant and EGFR-wild type tumours (p<0.001).Since CT is routinely used in lung cancer diagnosis, the deep learning model provides a non-invasive and easy-to-use method for EGFR mutation status prediction.


Asunto(s)
Adenocarcinoma del Pulmón/diagnóstico , Adenocarcinoma del Pulmón/genética , Aprendizaje Profundo , Mutación , Anciano , Biología Computacional , Receptores ErbB/genética , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Redes Neurales de la Computación , Curva ROC , Reproducibilidad de los Resultados , Estudios Retrospectivos , Tomografía Computarizada por Rayos X
13.
Bioinformatics ; 34(17): 3044-3046, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29668835

RESUMEN

Summary: DNA methylation is an important mechanism regulating gene transcription, and its role in carcinogenesis has been extensively studied. Hyper and hypomethylation of genes is a major mechanism of gene expression deregulation in a wide range of diseases. At the same time, high-throughput DNA methylation assays have been developed generating vast amounts of genome wide DNA methylation measurements. We developed MethylMix, an algorithm implemented in R to identify disease specific hyper and hypomethylated genes. Here we present a new version of MethylMix that automates the construction of DNA-methylation and gene expression datasets from The Cancer Genome Atlas (TCGA). More precisely, MethylMix 2.0 incorporates two major updates: the automated downloading of DNA methylation and gene expression datasets from TCGA and the automated preprocessing of such datasets: value imputation, batch correction and CpG sites clustering within each gene. The resulting datasets can subsequently be analyzed with MethylMix to identify transcriptionally predictive methylation states. We show that the Differential Methylation Values created by MethylMix can be used for cancer subtyping. Availability and implementation: MethylMix 2.0 was implemented as an R package and is available in bioconductor. https://www.bioconductor.org/packages/release/bioc/html/MethylMix.html.


Asunto(s)
Metilación de ADN , ADN/metabolismo , Algoritmos , Análisis por Conglomerados , Genoma , Humanos , Neoplasias/genética , Programas Informáticos
14.
Radiology ; 286(1): 307-315, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28727543

RESUMEN

Purpose To create a radiogenomic map linking computed tomographic (CT) image features and gene expression profiles generated by RNA sequencing for patients with non-small cell lung cancer (NSCLC). Materials and Methods A cohort of 113 patients with NSCLC diagnosed between April 2008 and September 2014 who had preoperative CT data and tumor tissue available was studied. For each tumor, a thoracic radiologist recorded 87 semantic image features, selected to reflect radiologic characteristics of nodule shape, margin, texture, tumor environment, and overall lung characteristics. Next, total RNA was extracted from the tissue and analyzed with RNA sequencing technology. Ten highly coexpressed gene clusters, termed metagenes, were identified, validated in publicly available gene-expression cohorts, and correlated with prognosis. Next, a radiogenomics map was built that linked semantic image features to metagenes by using the t statistic and the Spearman correlation metric with multiple testing correction. Results RNA sequencing analysis resulted in 10 metagenes that capture a variety of molecular pathways, including the epidermal growth factor (EGF) pathway. A radiogenomic map was created with 32 statistically significant correlations between semantic image features and metagenes. For example, nodule attenuation and margins are associated with the late cell-cycle genes, and a metagene that represents the EGF pathway was significantly correlated with the presence of ground-glass opacity and irregular nodules or nodules with poorly defined margins. Conclusion Radiogenomic analysis of NSCLC showed multiple associations between semantic image features and metagenes that represented canonical molecular pathways, and it can result in noninvasive identification of molecular properties of NSCLC. Online supplemental material is available for this article.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Genómica/métodos , Neoplasias Pulmonares , Imagen Molecular/métodos , Anciano , Anciano de 80 o más Años , Carcinoma de Pulmón de Células no Pequeñas/química , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/química , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/radioterapia , Masculino , Metagenoma , Persona de Mediana Edad , ARN Mensajero/análisis , ARN Mensajero/genética , Transducción de Señal
15.
J Neurooncol ; 140(2): 297-306, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30097822

RESUMEN

PURPOSE: To perform radiomics analysis for non-invasively predicting chromosome 1p/19q co-deletion in World Health Organization grade II and III (lower-grade) gliomas. METHODS: This retrospective study included 277 patients histopathologically diagnosed with lower-grade glioma. Clinical parameters were recorded for each patient. We performed a radiomics analysis by extracting 647 MRI-based features and applied the random forest algorithm to generate a radiomics signature for predicting 1p/19q co-deletion in the training cohort (n = 184). The clinical model consisted of pertinent clinical factors, and was built using a logistic regression algorithm. A combined model, incorporating both the radiomics signature and related clinical factors, was also constructed. The receiver operating characteristics curve was used to evaluate the predictive performance. We further validated the predictability of the three developed models using a time-independent validation cohort (n = 93). RESULTS: The radiomics signature was constructed as an independent predictor for differentiating 1p/19q co-deletion genotypes, which demonstrated superior performance on both the training and validation cohorts with areas under curve (AUCs) of 0.887 and 0.760, respectively. These results outperformed the clinical model (AUCs of 0.580 and 0.627 on training and validation cohorts). The AUCs of the combined model were 0.885 and 0.753 on training and validation cohorts, respectively, which indicated that clinical factors did not present additional improvement for the prediction. CONCLUSION: Our study highlighted that an MRI-based radiomics signature can effectively identify the 1p/19q co-deletion in histopathologically diagnosed lower-grade gliomas, thereby offering the potential to facilitate non-invasive molecular subtype prediction of gliomas.


Asunto(s)
Neoplasias Encefálicas/diagnóstico por imagen , Deleción Cromosómica , Cromosomas Humanos Par 19 , Cromosomas Humanos Par 1 , Glioma/diagnóstico por imagen , Imagen por Resonancia Magnética , Adolescente , Adulto , Anciano , Área Bajo la Curva , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Femenino , Glioma/genética , Glioma/patología , Humanos , Interpretación de Imagen Asistida por Computador/métodos , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Curva ROC , Estudios Retrospectivos , Adulto Joven
16.
BMC Bioinformatics ; 18(1): 32, 2017 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-28086747

RESUMEN

BACKGROUND: The current state-of-the-art in cancer diagnosis and treatment is not ideal; diagnostic tests are accurate but invasive, and treatments are "one-size fits-all" instead of being personalized. Recently, miRNA's have garnered significant attention as cancer biomarkers, owing to their ease of access (circulating miRNA in the blood) and stability. There have been many studies showing the effectiveness of miRNA data in diagnosing specific cancer types, but few studies explore the role of miRNA in predicting treatment outcome. METHODS: Here we go a step further, using tissue miRNA and clinical data across 21 cancers from the 'The Cancer Genome Atlas' (TCGA) database. We use machine learning techniques to create an accurate pan-cancer diagnosis system, and a prediction model for treatment outcomes. Finally, using these models, we create a web-based tool that diagnoses cancer and recommends the best treatment options. RESULTS: We achieved 97.2% accuracy for classification using a support vector machine classifier with radial basis. The accuracies improved to 99.9-100% when climbing up the embryonic tree and classifying cancers at different stages. We define the accuracy as the ratio of the total number of instances correctly classified to the total instances. The classifier also performed well, achieving greater than 80% sensitivity for many cancer types on independent validation datasets. Many miRNAs selected by our feature selection algorithm had strong previous associations to various cancers and tumor progression. Then, using miRNA, clinical and treatment data and encoding it in a machine-learning readable format, we built a prognosis predictor model to predict the outcome of treatment with 85% accuracy. We used this model to create a tool that recommends personalized treatment regimens. Both the diagnosis and prognosis model, incorporating semi-supervised learning techniques to improve their accuracies with repeated use, were uploaded online for easy access. CONCLUSION: Our research is a step towards the final goal of diagnosing cancer and predicting treatment recommendations using non-invasive blood tests.


Asunto(s)
MicroARNs/genética , Neoplasias/diagnóstico , Neoplasias/terapia , Algoritmos , Biomarcadores de Tumor/análisis , Humanos , Pronóstico , Máquina de Vectores de Soporte , Resultado del Tratamiento
17.
J Biomed Inform ; 72: 132-139, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28625880

RESUMEN

A crucial and limiting factor in data reuse is the lack of accurate, structured, and complete descriptions of data, known as metadata. Towards improving the quantity and quality of metadata, we propose a novel metadata prediction framework to learn associations from existing metadata that can be used to predict metadata values. We evaluate our framework in the context of experimental metadata from the Gene Expression Omnibus (GEO). We applied four rule mining algorithms to the most common structured metadata elements (sample type, molecular type, platform, label type and organism) from over 1.3million GEO records. We examined the quality of well supported rules from each algorithm and visualized the dependencies among metadata elements. Finally, we evaluated the performance of the algorithms in terms of accuracy, precision, recall, and F-measure. We found that PART is the best algorithm outperforming Apriori, Predictive Apriori, and Decision Table. All algorithms perform significantly better in predicting class values than the majority vote classifier. We found that the performance of the algorithms is related to the dimensionality of the GEO elements. The average performance of all algorithm increases due of the decreasing of dimensionality of the unique values of these elements (2697 platforms, 537 organisms, 454 labels, 9 molecules, and 5 types). Our work suggests that experimental metadata such as present in GEO can be accurately predicted using rule mining algorithms. Our work has implications for both prospective and retrospective augmentation of metadata quality, which are geared towards making data easier to find and reuse.


Asunto(s)
Algoritmos , Bases de Datos Genéticas , Expresión Génica , Metadatos , Humanos , Estudios Prospectivos , Estudios Retrospectivos
18.
Bioinformatics ; 31(11): 1839-41, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25609794

RESUMEN

UNLABELLED: DNA methylation is an important mechanism regulating gene transcription, and its role in carcinogenesis has been extensively studied. Hyper and hypomethylation of genes is an alternative mechanism to deregulate gene expression in a wide range of diseases. At the same time, high-throughput DNA methylation assays have been developed generating vast amounts of genome wide DNA methylation measurements. Yet, few tools exist that can formally identify hypo and hypermethylated genes that are predictive of transcription and thus functionally relevant for a particular disease. To accommodate this lack of tools, we developed MethylMix, an algorithm implemented in R to identify disease specific hyper and hypomethylated genes. MethylMix is based on a beta mixture model to identify methylation states and compares them with the normal DNA methylation state. MethylMix introduces a novel metric, the 'Differential Methylation value' or DM-value defined as the difference of a methylation state with the normal methylation state. Finally, matched gene expression data are used to identify, besides differential, transcriptionally predictive methylation states by focusing on methylation changes that effect gene expression. AVAILABILITY AND IMPLEMENTATION: MethylMix was implemented as an R package and is available in bioconductor.


Asunto(s)
Metilación de ADN , Enfermedad/genética , Regulación de la Expresión Génica , Programas Informáticos , Algoritmos , Humanos , Transcripción Genética
19.
BMC Cancer ; 16(1): 736, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27633254

RESUMEN

BACKGROUND: The incidence of Papillary thyroid carcinoma (PTC), the most common type of thyroid malignancy, has risen rapidly worldwide. PTC usually has an excellent prognosis. However, the rising incidence of PTC, due at least partially to widespread use of neck imaging studies with increased detection of small cancers, has created a clinical issue of overdiagnosis, and consequential overtreatment. We investigated how molecular data can be used to develop a prognostics signature for PTC. METHODS: The Cancer Genome Atlas (TCGA) recently reported on the genomic landscape of a large cohort of PTC cases. In order to decrease unnecessary morbidity associated with over diagnosing PTC patient with good prognosis, we used TCGA data to develop a gene expression signature to distinguish between patients with good and poor prognosis. We selected a set of clinical phenotypes to define an 'extreme poor' prognosis group and an 'extreme good' prognosis group and developed a gene signature that characterized these. RESULTS: We discovered a gene expression signature that distinguished the extreme good from extreme poor prognosis patients. Next, we applied this signature to the remaining intermediate risk patients, and show that they can be classified in clinically meaningful risk groups, characterized by established prognostic disease phenotypes. Analysis of the genes in the signature shows many known and novel genes involved in PTC prognosis. CONCLUSIONS: This work demonstrates that using a selection of clinical phenotypes and treatment variables, it is possible to develop a statistically useful and biologically meaningful gene signature of PTC prognosis, which may be developed as a biomarker to help prevent overdiagnosis.


Asunto(s)
Carcinoma/genética , Carcinoma/mortalidad , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/mortalidad , Transcriptoma , Adulto , Anciano , Biomarcadores de Tumor , Carcinoma/diagnóstico , Carcinoma Papilar , Análisis por Conglomerados , Biología Computacional/métodos , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Mutación , Metástasis de la Neoplasia , Estadificación de Neoplasias , Pronóstico , Timo/metabolismo , Cáncer Papilar Tiroideo , Neoplasias de la Tiroides/diagnóstico
20.
J Neuroradiol ; 42(4): 212-21, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24997477

RESUMEN

PURPOSE: The purpose of our study was to assess whether a model combining clinical factors, MR imaging features, and genomics would better predict overall survival of patients with glioblastoma (GBM) than either individual data type. METHODS: The study was conducted leveraging The Cancer Genome Atlas (TCGA) effort supported by the National Institutes of Health. Six neuroradiologists reviewed MRI images from The Cancer Imaging Archive (http://cancerimagingarchive.net) of 102 GBM patients using the VASARI scoring system. The patients' clinical and genetic data were obtained from the TCGA website (http://www.cancergenome.nih.gov/). Patient outcome was measured in terms of overall survival time. The association between different categories of biomarkers and survival was evaluated using Cox analysis. RESULTS: The features that were significantly associated with survival were: (1) clinical factors: chemotherapy; (2) imaging: proportion of tumor contrast enhancement on MRI; and (3) genomics: HRAS copy number variation. The combination of these three biomarkers resulted in an incremental increase in the strength of prediction of survival, with the model that included clinical, imaging, and genetic variables having the highest predictive accuracy (area under the curve 0.679±0.068, Akaike's information criterion 566.7, P<0.001). CONCLUSION: A combination of clinical factors, imaging features, and HRAS copy number variation best predicts survival of patients with GBM.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/mortalidad , Glioblastoma/diagnóstico , Glioblastoma/mortalidad , Imagen por Resonancia Magnética/métodos , Neoplasias Encefálicas/genética , Femenino , Marcadores Genéticos/genética , Predisposición Genética a la Enfermedad/epidemiología , Predisposición Genética a la Enfermedad/genética , Glioblastoma/genética , Humanos , Masculino , Prevalencia , Reproducibilidad de los Resultados , Estudios Retrospectivos , Medición de Riesgo/métodos , Sensibilidad y Especificidad , Análisis de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA