Svoboda | Graniru | BBC Russia | Golosameriki | Facebook
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,247)

Search Parameters:
Keywords = radiomics

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 4986 KiB  
Article
Radiomic Gradient in Peritumoural Tissue of Liver Metastases: A Biomarker for Clinical Practice? Analysing Density, Entropy, and Uniformity Variations with Distance from the Tumour
by Francesco Fiz, Elisa Maria Ragaini, Sara Sirchia, Chiara Masala, Samuele Viganò, Marco Francone, Lara Cavinato, Ettore Lanzarone, Angela Ammirabile and Luca Viganò
Diagnostics 2024, 14(14), 1552; https://doi.org/10.3390/diagnostics14141552 - 18 Jul 2024
Viewed by 166
Abstract
The radiomic analysis of the tissue surrounding colorectal liver metastases (CRLM) enhances the prediction accuracy of pathology data and survival. We explored the variation of the textural features in the peritumoural tissue as the distance from CRLM increases. We considered patients with hypodense [...] Read more.
The radiomic analysis of the tissue surrounding colorectal liver metastases (CRLM) enhances the prediction accuracy of pathology data and survival. We explored the variation of the textural features in the peritumoural tissue as the distance from CRLM increases. We considered patients with hypodense CRLMs >10 mm and high-quality computed tomography (CT). In the portal phase, we segmented (1) the tumour, (2) a series of concentric rims at a progressively increasing distance from CRLM (from one to ten millimetres), and (3) a cylinder of normal parenchyma (Liver-VOI). Sixty-three CRLMs in 51 patients were analysed. Median peritumoural HU values were similar to Liver-VOI, except for the first millimetre around the CRLM. Entropy progressively decreased (from 3.11 of CRLM to 2.54 of Liver-VOI), while uniformity increased (from 0.135 to 0.199, p < 0.001). At 10 mm from CRLM, entropy was similar to the Liver-VOI in 62% of cases and uniformity in 46%. In small CRLMs (≤30 mm) and responders to chemotherapy, normalisation of entropy and uniformity values occurred in a higher proportion of cases and at a shorter distance. The radiomic analysis of the parenchyma surrounding CRLMs unveiled a wide halo of progressively decreasing entropy and increasing uniformity despite a normal radiological aspect. Underlying pathology data should be investigated. Full article
Show Figures

Figure 1

14 pages, 4746 KiB  
Article
Differentiating Gliosarcoma from Glioblastoma: A Novel Approach Using PEACE and XGBoost to Deal with Datasets with Ultra-High Dimensional Confounders
by Amir Saki, Usef Faghihi and Ismaila Baldé
Life 2024, 14(7), 882; https://doi.org/10.3390/life14070882 - 16 Jul 2024
Viewed by 311
Abstract
In this study, we used a recently developed causal methodology, called Probabilistic Easy Variational Causal Effect (PEACE), to distinguish gliosarcoma (GSM) from glioblastoma (GBM). Our approach uses a causal metric which combines Probabilistic Easy Variational Causal Effect (PEACE) with the XGBoost, or eXtreme [...] Read more.
In this study, we used a recently developed causal methodology, called Probabilistic Easy Variational Causal Effect (PEACE), to distinguish gliosarcoma (GSM) from glioblastoma (GBM). Our approach uses a causal metric which combines Probabilistic Easy Variational Causal Effect (PEACE) with the XGBoost, or eXtreme Gradient Boosting, algorithm. Unlike prior research, which often relied on statistical models to reduce dataset dimensions before causal analysis, our approach uses the complete dataset with PEACE and the XGBoost algorithm. PEACE provides a comprehensive measurement of direct causal effects, applicable to both continuous and discrete variables. Our method provides both positive and negative versions of PEACE together with their averages to calculate the positive and negative causal effects of the radiomic features on the variable representing the type of tumor (GSM or GBM). In our model, PEACE and its variations are equipped with a degree d which varies from 0 to 1 and it reflects the importance of the rarity and frequency of the events. By using PEACE with XGBoost, we achieved a detailed and nuanced understanding of the causal relationships within the dataset features, facilitating accurate differentiation between GSM and GBM. To assess the XGBoost model, we used cross-validation and obtained a mean accuracy of 83% and an average model MSE of 0.130. This performance is notable given the high number of columns and low number of rows (code on GitHub). Full article
Show Figures

Figure 1

25 pages, 744 KiB  
Review
Understanding the Conundrum of Pancreatic Cancer in the Omics Sciences Era
by Alberto Nicoletti, Mattia Paratore, Federica Vitale, Marcantonio Negri, Giuseppe Quero, Giorgio Esposto, Irene Mignini, Sergio Alfieri, Antonio Gasbarrini, Maria Assunta Zocco and Lorenzo Zileri Dal Verme
Int. J. Mol. Sci. 2024, 25(14), 7623; https://doi.org/10.3390/ijms25147623 - 11 Jul 2024
Viewed by 356
Abstract
Pancreatic cancer (PC) is an increasing cause of cancer-related death, with a dismal prognosis caused by its aggressive biology, the lack of clinical symptoms in the early phases of the disease, and the inefficacy of treatments. PC is characterized by a complex tumor [...] Read more.
Pancreatic cancer (PC) is an increasing cause of cancer-related death, with a dismal prognosis caused by its aggressive biology, the lack of clinical symptoms in the early phases of the disease, and the inefficacy of treatments. PC is characterized by a complex tumor microenvironment. The interaction of its cellular components plays a crucial role in tumor development and progression, contributing to the alteration of metabolism and cellular hyperproliferation, as well as to metastatic evolution and abnormal tumor-associated immunity. Furthermore, in response to intrinsic oncogenic alterations and the influence of the tumor microenvironment, cancer cells undergo a complex oncogene-directed metabolic reprogramming that includes changes in glucose utilization, lipid and amino acid metabolism, redox balance, and activation of recycling and scavenging pathways. The advent of omics sciences is revolutionizing the comprehension of the pathogenetic conundrum of pancreatic carcinogenesis. In particular, metabolomics and genomics has led to a more precise classification of PC into subtypes that show different biological behaviors and responses to treatments. The identification of molecular targets through the pharmacogenomic approach may help to personalize treatments. Novel specific biomarkers have been discovered using proteomics and metabolomics analyses. Radiomics allows for an earlier diagnosis through the computational analysis of imaging. However, the complexity, high expertise required, and costs of the omics approach are the main limitations for its use in clinical practice at present. In addition, the studies of extracellular vesicles (EVs), the use of organoids, the understanding of host–microbiota interactions, and more recently the advent of artificial intelligence are helping to make further steps towards precision and personalized medicine. This present review summarizes the main evidence for the application of omics sciences to the study of PC and the identification of future perspectives. Full article
(This article belongs to the Special Issue Molecular Research on the Pancreatic Cancer)
Show Figures

Figure 1

13 pages, 2065 KiB  
Article
Investigation of Machine and Deep Learning Techniques to Detect HPV Status
by Efstathia Petrou, Konstantinos Chatzipapas, Panagiotis Papadimitroulas, Gustavo Andrade-Miranda, Paraskevi F. Katsakiori, Nikolaos D. Papathanasiou, Dimitris Visvikis and George C. Kagadis
J. Pers. Med. 2024, 14(7), 737; https://doi.org/10.3390/jpm14070737 - 10 Jul 2024
Viewed by 216
Abstract
Background: This study investigated alternative, non-invasive methods for human papillomavirus (HPV) detection in head and neck cancers (HNCs). We compared two approaches: analyzing computed tomography (CT) scans with a Deep Learning (DL) model and using radiomic features extracted from CT images with machine [...] Read more.
Background: This study investigated alternative, non-invasive methods for human papillomavirus (HPV) detection in head and neck cancers (HNCs). We compared two approaches: analyzing computed tomography (CT) scans with a Deep Learning (DL) model and using radiomic features extracted from CT images with machine learning (ML) models. Methods: Fifty patients with histologically confirmed HNC were included. We first trained a modified ResNet-18 DL model on CT data to predict HPV status. Next, radiomic features were extracted from manually segmented regions of interest near the oropharynx and used to train four ML models (K-Nearest Neighbors, logistic regression, decision tree, random forest) for the same purpose. Results: The CT-based model achieved the highest accuracy (90%) in classifying HPV status. Among the ML models, K-Nearest Neighbors performed best (80% accuracy). Weighted Ensemble methods combining the CT-based model with each ML model resulted in moderate accuracy improvements (70–90%). Conclusions: Our findings suggest that CT scans analyzed by DL models hold promise for non-invasive HPV detection in HNC. Radiomic features, while less accurate in this study, offer a complementary approach. Future research should explore larger datasets and investigate the potential of combining DL and radiomic techniques. Full article
Show Figures

Figure 1

9 pages, 787 KiB  
Brief Report
Enhanced Diagnostic Precision: Assessing Tumor Differentiation in Head and Neck Squamous Cell Carcinoma Using Multi-Slice Spiral CT Texture Analysis
by Lays Assolini Pinheiro de Oliveira, Diana Lorena Garcia Lopes, João Pedro Perez Gomes, Rafael Vinicius da Silveira, Daniel Vitor Aguiar Nozaki, Lana Ferreira Santos, Gabriela Castellano, Sérgio Lúcio Pereira de Castro Lopes and Andre Luiz Ferreira Costa
J. Clin. Med. 2024, 13(14), 4038; https://doi.org/10.3390/jcm13144038 - 10 Jul 2024
Viewed by 427
Abstract
This study explores the efficacy of texture analysis by using preoperative multi-slice spiral computed tomography (MSCT) to non-invasively determine the grade of cellular differentiation in head and neck squamous cell carcinoma (HNSCC). In a retrospective study, MSCT scans of patients with HNSCC were [...] Read more.
This study explores the efficacy of texture analysis by using preoperative multi-slice spiral computed tomography (MSCT) to non-invasively determine the grade of cellular differentiation in head and neck squamous cell carcinoma (HNSCC). In a retrospective study, MSCT scans of patients with HNSCC were analyzed and classified based on its histological grade as moderately differentiated, well-differentiated, or poorly differentiated. The location of the tumor was categorized as either in the bone or in soft tissues. Segmentation of the lesion areas was conducted, followed by texture analysis. Eleven GLCM parameters across five different distances were calculated. Median values and correlations of texture parameters were examined in relation to tumor differentiation grade by using Spearman’s correlation coefficient and Kruskal–Wallis and Dunn tests. Forty-six patients were included, predominantly female (87%), with a mean age of 66.7 years. Texture analysis revealed significant parameter correlations with histopathological grades of tumor differentiation. The study identified no significant age correlation with tumor differentiation, which underscores the potential of texture analysis as an age-independent biomarker. The strong correlations between texture parameters and histopathological grades support the integration of this technique into the clinical decision-making process. Full article
(This article belongs to the Topic AI in Medical Imaging and Image Processing)
Show Figures

Figure 1

13 pages, 268 KiB  
Review
The Role of Artificial Intelligence in Early Diagnosis and Molecular Classification of Head and Neck Skin Cancers: A Multidisciplinary Approach
by Zeliha Merve Semerci, Havva Serap Toru, Esra Çobankent Aytekin, Hümeyra Tercanlı, Diana Maria Chiorean, Yalçın Albayrak and Ovidiu Simion Cotoi
Diagnostics 2024, 14(14), 1477; https://doi.org/10.3390/diagnostics14141477 - 10 Jul 2024
Viewed by 284
Abstract
Cancer remains a significant global health concern, with increasing genetic and metabolic irregularities linked to its onset. Among various forms of cancer, skin cancer, including squamous cell carcinoma, basal cell carcinoma, and melanoma, is on the rise worldwide, often triggered by ultraviolet (UV) [...] Read more.
Cancer remains a significant global health concern, with increasing genetic and metabolic irregularities linked to its onset. Among various forms of cancer, skin cancer, including squamous cell carcinoma, basal cell carcinoma, and melanoma, is on the rise worldwide, often triggered by ultraviolet (UV) radiation. The propensity of skin cancer to metastasize highlights the importance of early detection for successful treatment. This narrative review explores the evolving role of artificial intelligence (AI) in diagnosing head and neck skin cancers from both radiological and pathological perspectives. In the past two decades, AI has made remarkable progress in skin cancer research, driven by advances in computational capabilities, digitalization of medical images, and radiomics data. AI has shown significant promise in image-based diagnosis across various medical domains. In dermatology, AI has played a pivotal role in refining diagnostic and treatment strategies, including genomic risk assessment. This technology offers substantial potential to aid primary clinicians in improving patient outcomes. Studies have demonstrated AI’s effectiveness in identifying skin lesions, categorizing them, and assessing their malignancy, contributing to earlier interventions and better prognosis. The rising incidence and mortality rates of skin cancer, coupled with the high cost of treatment, emphasize the need for early diagnosis. Further research and integration of AI into clinical practice are warranted to maximize its benefits in skin cancer diagnosis and treatment. Full article
(This article belongs to the Section Pathology and Molecular Diagnostics)
22 pages, 3817 KiB  
Article
Enhancing Immunotherapy Response Prediction in Metastatic Lung Adenocarcinoma: Leveraging Shallow and Deep Learning with CT-Based Radiomics across Single and Multiple Tumor Sites
by Cécile Masson-Grehaigne, Mathilde Lafon, Jean Palussière, Laura Leroy, Benjamin Bonhomme, Eva Jambon, Antoine Italiano, Sophie Cousin and Amandine Crombé
Cancers 2024, 16(13), 2491; https://doi.org/10.3390/cancers16132491 - 8 Jul 2024
Viewed by 485
Abstract
This study aimed to evaluate the potential of pre-treatment CT-based radiomics features (RFs) derived from single and multiple tumor sites, and state-of-the-art machine-learning survival algorithms, in predicting progression-free survival (PFS) for patients with metastatic lung adenocarcinoma (MLUAD) receiving first-line treatment including immune checkpoint [...] Read more.
This study aimed to evaluate the potential of pre-treatment CT-based radiomics features (RFs) derived from single and multiple tumor sites, and state-of-the-art machine-learning survival algorithms, in predicting progression-free survival (PFS) for patients with metastatic lung adenocarcinoma (MLUAD) receiving first-line treatment including immune checkpoint inhibitors (CPIs). To do so, all adults with newly diagnosed MLUAD, pre-treatment contrast-enhanced CT scan, and performance status ≤ 2 who were treated at our cancer center with first-line CPI between November 2016 and November 2022 were included. RFs were extracted from all measurable lesions with a volume ≥ 1 cm3 on the CT scan. To capture intra- and inter-tumor heterogeneity, RFs from the largest tumor of each patient, as well as lowest, highest, and average RF values over all lesions per patient were collected. Intra-patient inter-tumor heterogeneity metrics were calculated to measure the similarity between each patient lesions. After filtering predictors with univariable Cox p < 0.100 and analyzing their correlations, five survival machine-learning algorithms (stepwise Cox regression [SCR], LASSO Cox regression, random survival forests, gradient boosted machine [GBM], and deep learning [Deepsurv]) were trained in 100-times repeated 5-fold cross-validation (rCV) to predict PFS on three inputs: (i) clinicopathological variables, (ii) all radiomics-based and clinicopathological (full input), and (iii) uncorrelated radiomics-based and clinicopathological variables (uncorrelated input). The Models’ performances were evaluated using the concordance index (c-index). Overall, 140 patients were included (median age: 62.5 years, 36.4% women). In rCV, the highest c-index was reached with Deepsurv (c-index = 0.631, 95%CI = 0.625–0.647), followed by GBM (c-index = 0.603, 95%CI = 0.557–0.646), significantly outperforming standard SCR whatever its input (c-index range: 0.560–0.570, all p < 0.0001). Thus, single- and multi-site pre-treatment radiomics data provide valuable prognostic information for predicting PFS in MLUAD patients undergoing first-line CPI treatment when analyzed with advanced machine-learning survival algorithms. Full article
(This article belongs to the Special Issue Imaging and Molecular Biology as Biomarkers for Lung Cancer)
Show Figures

Figure 1

11 pages, 1085 KiB  
Review
Advancements in Radiogenomics for Clear Cell Renal Cell Carcinoma: Understanding the Impact of BAP1 Mutation
by Federico Greco, Valerio D’Andrea, Andrea Buoso, Laura Cea, Caterina Bernetti, Bruno Beomonte Zobel and Carlo Augusto Mallio
J. Clin. Med. 2024, 13(13), 3960; https://doi.org/10.3390/jcm13133960 - 6 Jul 2024
Viewed by 454
Abstract
Recent advancements in understanding clear cell renal cell carcinoma (ccRCC) have underscored the critical role of the BAP1 gene in its pathogenesis and prognosis. While the von Hippel–Lindau (VHL) mutation has been extensively studied, emerging evidence suggests that mutations in BAP1 and other [...] Read more.
Recent advancements in understanding clear cell renal cell carcinoma (ccRCC) have underscored the critical role of the BAP1 gene in its pathogenesis and prognosis. While the von Hippel–Lindau (VHL) mutation has been extensively studied, emerging evidence suggests that mutations in BAP1 and other genes significantly impact patient outcomes. Radiogenomics with and without texture analysis based on CT imaging holds promise in predicting BAP1 mutation status and overall survival outcomes. However, prospective studies with larger cohorts and standardized imaging protocols are needed to validate these findings and translate them into clinical practice effectively, paving the way for personalized treatment strategies in ccRCC. This review aims to summarize the current knowledge on the role of BAP1 mutation in ccRCC pathogenesis and prognosis, as well as the potential of radiogenomics in predicting mutation status and clinical outcomes. Full article
(This article belongs to the Special Issue Advanced Imaging Techniques for Nephrology and Urology)
Show Figures

Figure 1

12 pages, 9083 KiB  
Article
Prediction of the Benign or Malignant Nature of Pulmonary Pure Ground-Glass Nodules Based on Radiomics Analysis of High-Resolution Computed Tomography Images
by Xiaoxia Ping, Nan Jiang, Qian Meng and Chunhong Hu
Tomography 2024, 10(7), 1042-1053; https://doi.org/10.3390/tomography10070078 - 5 Jul 2024
Viewed by 228
Abstract
To evaluate the efficacy of radiomics features extracted from preoperative high-resolution computed tomography (HRCT) scans in distinguishing benign and malignant pulmonary pure ground-glass nodules (pGGNs), a retrospective study of 395 patients from 2016 to 2020 was conducted. All nodules were randomly divided into [...] Read more.
To evaluate the efficacy of radiomics features extracted from preoperative high-resolution computed tomography (HRCT) scans in distinguishing benign and malignant pulmonary pure ground-glass nodules (pGGNs), a retrospective study of 395 patients from 2016 to 2020 was conducted. All nodules were randomly divided into the training and validation sets in the ratio of 7:3. Radiomics features were extracted using MaZda software (version 4.6), and the least absolute shrinkage and selection operator (LASSO) was employed for feature selection. Significant differences were observed in the training set between benign and malignant pGGNs in sex, mean CT value, margin, pleural retraction, tumor–lung interface, and internal vascular change, and then the mean CT value and the morphological features model were constructed. Fourteen radiomics features were selected by LASSO for the radiomics model. The combined model was developed by integrating all selected radiographic and radiomics features using logistic regression. The AUCs in the training set were 0.606 for the mean CT value, 0.718 for morphological features, 0.756 for radiomics features, and 0.808 for the combined model. In the validation set, AUCs were 0.601, 0.692, 0.696, and 0.738, respectively. The decision curves showed that the combined model demonstrated the highest net benefit. Full article
Show Figures

Figure 1

12 pages, 745 KiB  
Article
Radiomics-Guided Deep Learning Networks Classify Differential Diagnosis of Parkinsonism
by Ronghua Ling, Min Wang, Jiaying Lu, Shaoyou Wu, Ping Wu, Jingjie Ge, Luyao Wang, Yingqian Liu, Juanjuan Jiang, Kuangyu Shi, Zhuangzhi Yan, Chuantao Zuo and Jiehui Jiang
Brain Sci. 2024, 14(7), 680; https://doi.org/10.3390/brainsci14070680 - 4 Jul 2024
Viewed by 664
Abstract
The differential diagnosis between atypical Parkinsonian syndromes may be challenging and critical. We aimed to proposed a radiomics-guided deep learning (DL) model to discover interpretable DL features and further verify the proposed model through the differential diagnosis of Parkinsonian syndromes. We recruited 1495 [...] Read more.
The differential diagnosis between atypical Parkinsonian syndromes may be challenging and critical. We aimed to proposed a radiomics-guided deep learning (DL) model to discover interpretable DL features and further verify the proposed model through the differential diagnosis of Parkinsonian syndromes. We recruited 1495 subjects for 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET) scanning, including 220 healthy controls and 1275 patients diagnosed with idiopathic Parkinson’s disease (IPD), multiple system atrophy (MSA), or progressive supranuclear palsy (PSP). Baseline radiomics and two DL models were developed and tested for the Parkinsonian diagnosis. The DL latent features were extracted from the last layer and subsequently guided by radiomics. The radiomics-guided DL model outperformed the baseline radiomics approach, suggesting the effectiveness of the DL approach. DenseNet showed the best diagnosis ability (sensitivity: 95.7%, 90.1%, and 91.2% for IPD, MSA, and PSP, respectively) using retained DL features in the test dataset. The retained DL latent features were significantly associated with radiomics features and could be interpreted through biological explanations of handcrafted radiomics features. The radiomics-guided DL model offers interpretable high-level abstract information for differential diagnosis of Parkinsonian disorders and holds considerable promise for personalized disease monitoring. Full article
(This article belongs to the Special Issue Challenges and Perspectives of Neurological Disorders: Series II)
Show Figures

Figure 1

18 pages, 2135 KiB  
Article
Stability of Radiomic Features against Variations in Lesion Segmentations Computed on Apparent Diffusion Coefficient Maps of Breast Lesions
by Mona Pistel, Luise Brock, Frederik Bernd Laun, Ramona Erber, Elisabeth Weiland, Michael Uder, Evelyn Wenkel, Sabine Ohlmeyer and Sebastian Bickelhaupt
Diagnostics 2024, 14(13), 1427; https://doi.org/10.3390/diagnostics14131427 - 3 Jul 2024
Viewed by 442
Abstract
Diffusion-weighted imaging (DWI) combined with radiomics can aid in the differentiation of breast lesions. Segmentation characteristics, however, might influence radiomic features. To evaluate feature stability, we implemented a standardized pipeline featuring shifts and shape variations of the underlying segmentations. A total of 103 [...] Read more.
Diffusion-weighted imaging (DWI) combined with radiomics can aid in the differentiation of breast lesions. Segmentation characteristics, however, might influence radiomic features. To evaluate feature stability, we implemented a standardized pipeline featuring shifts and shape variations of the underlying segmentations. A total of 103 patients were retrospectively included in this IRB-approved study after multiparametric diagnostic breast 3T MRI with a spin-echo diffusion-weighted sequence with echoplanar readout (b-values: 50, 750 and 1500 s/mm2). Lesion segmentations underwent shifts and shape variations, with >100 radiomic features extracted from apparent diffusion coefficient (ADC) maps for each variation. These features were then compared and ranked based on their stability, measured by the Overall Concordance Correlation Coefficient (OCCC) and Dynamic Range (DR). Results showed variation in feature robustness to segmentation changes. The most stable features, excluding shape-related features, were FO (Mean, Median, RootMeanSquared), GLDM (DependenceNonUniformity), GLRLM (RunLengthNonUniformity), and GLSZM (SizeZoneNonUniformity), which all had OCCC and DR > 0.95 for both shifting and resizing the segmentation. Perimeter, MajorAxisLength, MaximumDiameter, PixelSurface, MeshSurface, and MinorAxisLength were the most stable features in the Shape category with OCCC and DR > 0.95 for resizing. Considering the variability in radiomic feature stability against segmentation variations is relevant when interpreting radiomic analysis of breast DWI data. Full article
(This article belongs to the Section Medical Imaging and Theranostics)
Show Figures

Figure 1

24 pages, 1573 KiB  
Review
Integrating Omics Data and AI for Cancer Diagnosis and Prognosis
by Yousaku Ozaki, Phil Broughton, Hamed Abdollahi, Homayoun Valafar and Anna V. Blenda
Cancers 2024, 16(13), 2448; https://doi.org/10.3390/cancers16132448 - 3 Jul 2024
Viewed by 888
Abstract
Cancer is one of the leading causes of death, making timely diagnosis and prognosis very important. Utilization of AI (artificial intelligence) enables providers to organize and process patient data in a way that can lead to better overall outcomes. This review paper aims [...] Read more.
Cancer is one of the leading causes of death, making timely diagnosis and prognosis very important. Utilization of AI (artificial intelligence) enables providers to organize and process patient data in a way that can lead to better overall outcomes. This review paper aims to look at the varying uses of AI for diagnosis and prognosis and clinical utility. PubMed and EBSCO databases were utilized for finding publications from 1 January 2020 to 22 December 2023. Articles were collected using key search terms such as “artificial intelligence” and “machine learning.” Included in the collection were studies of the application of AI in determining cancer diagnosis and prognosis using multi-omics data, radiomics, pathomics, and clinical and laboratory data. The resulting 89 studies were categorized into eight sections based on the type of data utilized and then further subdivided into two subsections focusing on cancer diagnosis and prognosis, respectively. Eight studies integrated more than one form of omics, namely genomics, transcriptomics, epigenomics, and proteomics. Incorporating AI into cancer diagnosis and prognosis alongside omics and clinical data represents a significant advancement. Given the considerable potential of AI in this domain, ongoing prospective studies are essential to enhance algorithm interpretability and to ensure safe clinical integration. Full article
(This article belongs to the Special Issue Oncogenomic and Multi-Omic Data Science and Data Engineering)
Show Figures

Figure 1

20 pages, 569 KiB  
Systematic Review
Radiomic Pipelines for Prostate Cancer in External Beam Radiation Therapy: A Review of Methods and Future Directions
by Bruno Mendes, Inês Domingues and João Santos
J. Clin. Med. 2024, 13(13), 3907; https://doi.org/10.3390/jcm13133907 - 3 Jul 2024
Viewed by 460
Abstract
Background: Prostate Cancer (PCa) is asymptomatic at an early stage and often painless, requiring only active surveillance. External Beam Radiotherapy (EBRT) is currently a curative option for localised and locally advanced diseases and a palliative option for metastatic low-volume disease. Although highly [...] Read more.
Background: Prostate Cancer (PCa) is asymptomatic at an early stage and often painless, requiring only active surveillance. External Beam Radiotherapy (EBRT) is currently a curative option for localised and locally advanced diseases and a palliative option for metastatic low-volume disease. Although highly effective, especially in a hypofractionation scheme, 17.4% to 39.4% of all patients suffer from cancer recurrence after EBRT. But, radiographic findings also correlate with significant differences in protein expression patterns. In the PCa EBRT workflow, several imaging modalities are available for grading, staging and contouring. Using image data characterisation algorithms (radiomics), one can provide a quantitative analysis of prognostic and predictive treatment outcomes. Methods: This literature review searched for original studies in radiomics for PCa in the context of EBRT. Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, this review includes 73 new studies and analyses datasets, imaging modality, segmentation technique, feature extraction, selection and model building methods. Results: Magnetic Resonance Imaging (MRI) is the preferred imaging modality for radiomic studies in PCa but Computed Tomography (CT), Positron Emission Tomography (PET) and Ultrasound (US) may offer valuable insights on tumour characterisation and treatment response prediction. Conclusions: Most radiomic studies used small, homogeneous and private datasets lacking external validation and variability. Future research should focus on collaborative efforts to create large, multicentric datasets and develop standardised methodologies, ensuring the full potential of radiomics in clinical practice. Full article
(This article belongs to the Section Nephrology & Urology)
Show Figures

Figure 1

34 pages, 3635 KiB  
Article
Machine Learning for Extraction of Image Features Associated with Progression of Geographic Atrophy
by Janan Arslan and Kurt Benke
BioMedInformatics 2024, 4(3), 1638-1671; https://doi.org/10.3390/biomedinformatics4030089 - 2 Jul 2024
Viewed by 350
Abstract
Background: Several studies have investigated various features and models in order to understand the growth and progression of the ocular disease geographic atrophy (GA). Commonly assessed features include age, sex, smoking, alcohol consumption, sedentary lifestyle, hypertension, and diabetes. There have been inconsistencies regarding [...] Read more.
Background: Several studies have investigated various features and models in order to understand the growth and progression of the ocular disease geographic atrophy (GA). Commonly assessed features include age, sex, smoking, alcohol consumption, sedentary lifestyle, hypertension, and diabetes. There have been inconsistencies regarding which features correlate with GA progression. Chief amongst these inconsistencies is whether the investigated features are readily available for analysis across various ophthalmic institutions. Methods:In this study, we focused our attention on the association of fundus autofluorescence (FAF) imaging features and GA progression. Our method included feature extraction using radiomic processes and feature ranking by machine learning incorporating the algorithm XGBoost to determine the best-ranked features. This led to the development of an image-based linear mixed-effects model, which was designed to account for slope change based on within-subject variability and inter-eye correlation. Metrics used to assess the linear mixed-effects model included marginal and conditional R2, Pearson’s correlation coefficient (r), root mean square error (RMSE), mean error (ME), mean absolute error (MAE), mean absolute deviation (MAD), the Akaike Information Criterion (AIC), the Bayesian Information Criterion (BIC), and loglikelihood. Results: We developed a linear mixed-effects model with 15 image-based features. The model results were as follows: R2 = 0.96, r = 0.981, RMSE = 1.32, ME = −7.3 × 10−15, MAE = 0.94, MAD = 0.999, AIC = 2084.93, BIC = 2169.97, and log likelihood = −1022.46. Conclusions: The advantage of our method is that it relies on the inherent properties of the image itself, rather than the availability of clinical or demographic data. Thus, the image features discovered in this study are universally and readily available across the board. Full article
(This article belongs to the Special Issue Editor's Choices Series for Methods in Biomedical Informatics Section)
Show Figures

Figure 1

17 pages, 3526 KiB  
Article
Outcome Prediction Using Multi-Modal Information: Integrating Large Language Model-Extracted Clinical Information and Image Analysis
by Di Sun, Lubomir Hadjiiski, John Gormley, Heang-Ping Chan, Elaine Caoili, Richard Cohan, Ajjai Alva, Grace Bruno, Rada Mihalcea, Chuan Zhou and Vikas Gulani
Cancers 2024, 16(13), 2402; https://doi.org/10.3390/cancers16132402 - 29 Jun 2024
Viewed by 496
Abstract
Survival prediction post-cystectomy is essential for the follow-up care of bladder cancer patients. This study aimed to evaluate artificial intelligence (AI)-large language models (LLMs) for extracting clinical information and improving image analysis, with an initial application involving predicting five-year survival rates of patients [...] Read more.
Survival prediction post-cystectomy is essential for the follow-up care of bladder cancer patients. This study aimed to evaluate artificial intelligence (AI)-large language models (LLMs) for extracting clinical information and improving image analysis, with an initial application involving predicting five-year survival rates of patients after radical cystectomy for bladder cancer. Data were retrospectively collected from medical records and CT urograms (CTUs) of bladder cancer patients between 2001 and 2020. Of 781 patients, 163 underwent chemotherapy, had pre- and post-chemotherapy CTUs, underwent radical cystectomy, and had an available post-surgery five-year survival follow-up. Five AI-LLMs (Dolly-v2, Vicuna-13b, Llama-2.0-13b, GPT-3.5, and GPT-4.0) were used to extract clinical descriptors from each patient’s medical records. As a reference standard, clinical descriptors were also extracted manually. Radiomics and deep learning descriptors were extracted from CTU images. The developed multi-modal predictive model, CRD, was based on the clinical (C), radiomics (R), and deep learning (D) descriptors. The LLM retrieval accuracy was assessed. The performances of the survival predictive models were evaluated using AUC and Kaplan–Meier analysis. For the 163 patients (mean age 64 ± 9 years; M:F 131:32), the LLMs achieved extraction accuracies of 74%~87% (Dolly), 76%~83% (Vicuna), 82%~93% (Llama), 85%~91% (GPT-3.5), and 94%~97% (GPT-4.0). For a test dataset of 64 patients, the CRD model achieved AUCs of 0.89 ± 0.04 (manually extracted information), 0.87 ± 0.05 (Dolly), 0.83 ± 0.06~0.84 ± 0.05 (Vicuna), 0.81 ± 0.06~0.86 ± 0.05 (Llama), 0.85 ± 0.05~0.88 ± 0.05 (GPT-3.5), and 0.87 ± 0.05~0.88 ± 0.05 (GPT-4.0). This study demonstrates the use of LLM model-extracted clinical information, in conjunction with imaging analysis, to improve the prediction of clinical outcomes, with bladder cancer as an initial example. Full article
Show Figures

Figure 1

Back to TopTop