Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters











Database
Language
Publication year range
1.
J Pers Med ; 14(5)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38793065

ABSTRACT

Radiotherapy is focused on the tumor but also reaches healthy tissues, causing toxicities that are possibly related to genomic factors. In this context, radiogenomics can help reduce the toxicity, increase the effectiveness of radiotherapy, and personalize treatment. It is important to consider the genomic profiles of populations not yet studied in radiogenomics, such as the indigenous Amazonian population. Thus, our objective was to analyze important genes for radiogenomics, such as ATM, TGFB1, RAD51, AREG, XRCC4, CDK1, MEG3, PRKCE, TANC1, and KDR, in indigenous people and draw a radiogenomic profile of this population. The NextSeq 500® platform was used for sequencing reactions; for differences in the allelic frequency between populations, Fisher's Exact Test was used. We identified 39 variants, 2 of which were high impact: 1 in KDR (rs41452948) and another in XRCC4 (rs1805377). We found four modifying variants not yet described in the literature in PRKCE. We did not find any variants in TANC1-an important gene for personalized medicine in radiotherapy-that were associated with toxicities in previous cohorts, configuring a protective factor for indigenous people. We identified four SNVs (rs664143, rs1801516, rs1870377, rs1800470) that were associated with toxicity in previous studies. Knowing the radiogenomic profile of indigenous people can help personalize their radiotherapy.

2.
BMC Bioinformatics ; 24(1): 401, 2023 Oct 26.
Article in English | MEDLINE | ID: mdl-37884877

ABSTRACT

BACKGROUND: Recent advancements in computing power and state-of-the-art algorithms have helped in more accessible and accurate diagnosis of numerous diseases. In addition, the development of de novo areas in imaging science, such as radiomics and radiogenomics, have been adding more to personalize healthcare to stratify patients better. These techniques associate imaging phenotypes with the related disease genes. Various imaging modalities have been used for years to diagnose breast cancer. Nonetheless, digital breast tomosynthesis (DBT), a state-of-the-art technique, has produced promising results comparatively. DBT, a 3D mammography, is replacing conventional 2D mammography rapidly. This technological advancement is key to AI algorithms for accurately interpreting medical images. OBJECTIVE AND METHODS: This paper presents a comprehensive review of deep learning (DL), radiomics and radiogenomics in breast image analysis. This review focuses on DBT, its extracted synthetic mammography (SM), and full-field digital mammography (FFDM). Furthermore, this survey provides systematic knowledge about DL, radiomics, and radiogenomics for beginners and advanced-level researchers. RESULTS: A total of 500 articles were identified, with 30 studies included as the set criteria. Parallel benchmarking of radiomics, radiogenomics, and DL models applied to the DBT images could allow clinicians and researchers alike to have greater awareness as they consider clinical deployment or development of new models. This review provides a comprehensive guide to understanding the current state of early breast cancer detection using DBT images. CONCLUSION: Using this survey, investigators with various backgrounds can easily seek interdisciplinary science and new DL, radiomics, and radiogenomics directions towards DBT.


Subject(s)
Breast Neoplasms , Deep Learning , Humans , Female , Radiographic Image Enhancement/methods , Breast/diagnostic imaging , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/genetics , Mammography/methods
3.
Clin Imaging ; 84: 54-60, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35144039

ABSTRACT

With the rise of artificial intelligence, radiomics has emerged as a field of translational research based on the extraction of mineable high-dimensional data from radiological images to create "big data" datasets for the purpose of identifying distinct sub-visual imaging patterns. The integrated analysis of radiomic data and genomic data is termed radiogenomics, a promising strategy to identify potential imaging biomarkers for predicting driver mutations and other genomic parameters. In lung cancer, recent advances in whole-genome sequencing and the identification of actionable molecular alterations have led to an increased interest in understanding the complex relationships between imaging and genomic data, with the potential of guiding therapeutic strategies and predicting clinical outcomes. Although the integration of the radiogenomics data into lung cancer management may represent a new paradigm in the field, the use of this technique as a clinical biomarker remains investigational and still necessitates standardization and robustness to be effectively translated into the clinical practice. This review summarizes the basic concepts, potential contributions, challenges, and opportunities of radiogenomics in the management of patients with lung cancer.


Subject(s)
Lung Neoplasms , Radiology , Artificial Intelligence , Diagnostic Imaging , Genomics/methods , Humans , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/genetics , Lung Neoplasms/therapy
4.
Tomography ; 7(2): 154-168, 2021 04 29.
Article in English | MEDLINE | ID: mdl-33946756

ABSTRACT

Lung cancer causes more deaths globally than any other type of cancer. To determine the best treatment, detecting EGFR and KRAS mutations is of interest. However, non-invasive ways to obtain this information are not available. Furthermore, many times there is a lack of big enough relevant public datasets, so the performance of single classifiers is not outstanding. In this paper, an ensemble approach is applied to increase the performance of EGFR and KRAS mutation prediction using a small dataset. A new voting scheme, Selective Class Average Voting (SCAV), is proposed and its performance is assessed both for machine learning models and CNNs. For the EGFR mutation, in the machine learning approach, there was an increase in the sensitivity from 0.66 to 0.75, and an increase in AUC from 0.68 to 0.70. With the deep learning approach, an AUC of 0.846 was obtained, and with SCAV, the accuracy of the model was increased from 0.80 to 0.857. For the KRAS mutation, both in the machine learning models (0.65 to 0.71 AUC) and the deep learning models (0.739 to 0.778 AUC), a significant increase in performance was found. The results obtained in this work show how to effectively learn from small image datasets to predict EGFR and KRAS mutations, and that using ensembles with SCAV increases the performance of machine learning classifiers and CNNs. The results provide confidence that as large datasets become available, tools to augment clinical capabilities can be fielded.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/genetics , Humans , Lung Neoplasms/genetics , Mutation , Proto-Oncogene Proteins p21(ras)/genetics
5.
Abdom Radiol (NY) ; 44(11): 3764-3774, 2019 11.
Article in English | MEDLINE | ID: mdl-31055615

ABSTRACT

INTRODUCTION: As computational capabilities have advanced, radiologists and their collaborators have looked for novel ways to analyze diagnostic images. This has resulted in the development of radiomics and radiogenomics as new fields in medical imaging. Radiomics and radiogenomics may change the practice of medicine, particularly for patients with colorectal cancer. Radiomics corresponds to the extraction and analysis of numerous quantitative imaging features from conventional imaging modalities in correlation with several endpoints, including the prediction of pathology, genomics, therapeutic response, and clinical outcome. In radiogenomics, qualitative and/or quantitative imaging features are extracted and correlated with genetic profiles of the imaged tissue. Thus far, several studies have evaluated the use of radiomics and radiogenomics in patients with colorectal cancer; however, there are challenges to be overcome before its routine implementation including challenges related to sample size, model design and interpretability, and the lack of robust multicenter validation set. MATERIAL AND METHODS: In this article, we will review the concepts of radiomics and radiogenomics and their potential applications in rectal cancer. CONCLUSION: Radiologists should be aware of the basic concepts, benefits, pitfalls, and limitations of new radiomic and radiogenomics techniques to achieve a balanced interpretation of the results.


Subject(s)
Diagnostic Imaging/trends , Genomics , Image Interpretation, Computer-Assisted/methods , Rectal Neoplasms/diagnostic imaging , Rectal Neoplasms/genetics , Humans
SELECTION OF CITATIONS
SEARCH DETAIL