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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (767)

Search Parameters:
Keywords = cancer radiomics

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
13 pages, 1488 KiB  
Article
Validation of a Quantitative Ultrasound Texture Analysis Model for Early Prediction of Neoadjuvant Chemotherapy Response in Breast Cancer: A Prospective Serial Imaging Study
by Daniel Moore-Palhares, Lakshmanan Sannachi, Adrian Wai Chan, Archya Dasgupta, Daniel DiCenzo, Sonal Gandhi, Rossanna Pezo, Andrea Eisen, Ellen Warner, Frances Wright, Nicole Look Hong, Ali Sadeghi-Naini, Mia Skarpathiotakis, Belinda Curpen, Carrie Betel, Michael C. Kolios, Maureen Trudeau and Gregory J. Czarnota
Cancers 2025, 17(15), 2594; https://doi.org/10.3390/cancers17152594 - 7 Aug 2025
Abstract
Background/Objectives: Patients with breast cancer who do not achieve a complete response to neoadjuvant chemotherapy (NAC) may benefit from intensified adjuvant systemic therapy. However, such treatment escalation is typically delayed until after tumour resection, which occurs several months into the treatment course. Quantitative [...] Read more.
Background/Objectives: Patients with breast cancer who do not achieve a complete response to neoadjuvant chemotherapy (NAC) may benefit from intensified adjuvant systemic therapy. However, such treatment escalation is typically delayed until after tumour resection, which occurs several months into the treatment course. Quantitative ultrasound (QUS) can detect early microstructural changes in tumours and may enable timely identification of non-responders during NAC, allowing for earlier treatment intensification. In our previous prospective observational study, 100 breast cancer patients underwent QUS imaging before and four times during NAC. Machine learning algorithms based on QUS texture features acquired in the first week of treatment were developed and achieved 78% accuracy in predicting treatment response. In the current study, we aimed to validate these algorithms in an independent prospective cohort to assess reproducibility and confirm their clinical utility. Methods: We included breast cancer patients eligible for NAC per standard of care, with tumours larger than 1.5 cm. QUS imaging was acquired at baseline and during the first week of treatment. Tumour response was defined as a ≥30% reduction in target lesion size on the resection specimen compared to baseline imaging. Results: A total of 51 patients treated between 2018 and 2021 were included (median age 49 years; median tumour size 3.6 cm). Most were estrogen receptor–positive (65%) or HER2-positive (33%), and the majority received dose-dense AC-T (n = 34, 67%) or FEC-D (n = 15, 29%) chemotherapy, with or without trastuzumab. The support vector machine algorithm achieved an area under the curve of 0.71, with 86% accuracy, 91% specificity, 50% sensitivity, 93% negative predictive value, and 43% positive predictive value for predicting treatment response. Misclassifications were primarily associated with poorly defined tumours and difficulties in accurately identifying the region of interest. Conclusions: Our findings validate QUS-based machine learning models for early prediction of chemotherapy response and support their potential as non-invasive tools for treatment personalization and clinical trial development focused on early treatment intensification. Full article
(This article belongs to the Special Issue Clinical Applications of Ultrasound in Cancer Imaging and Treatment)
Show Figures

Figure 1

14 pages, 1848 KiB  
Article
RadiomiX for Radiomics Analysis: Automated Approaches to Overcome Challenges in Replicability
by Harel Kotler, Luca Bergamin, Fabio Aiolli, Elena Scagliori, Angela Grassi, Giulia Pasello, Alessandra Ferro, Francesca Caumo and Gisella Gennaro
Diagnostics 2025, 15(15), 1968; https://doi.org/10.3390/diagnostics15151968 - 5 Aug 2025
Abstract
Background/Objectives: To simplify the decision-making process in radiomics by employing RadiomiX, an algorithm designed to automatically identify the best model combination and validate them across multiple environments was developed, thus enhancing the reliability of results. Methods: RadiomiX systematically tests classifier and feature [...] Read more.
Background/Objectives: To simplify the decision-making process in radiomics by employing RadiomiX, an algorithm designed to automatically identify the best model combination and validate them across multiple environments was developed, thus enhancing the reliability of results. Methods: RadiomiX systematically tests classifier and feature selection method combinations known to be suitable for radiomic datasets to determine the best-performing configuration across multiple train–test splits and K-fold cross-validation. The framework was validated on four public retrospective radiomics datasets including lung nodules, metastatic breast cancer, and hepatic encephalopathy using CT, PET/CT, and MRI modalities. Model performance was assessed using the area under the receiver-operating-characteristic curve (AUC) and accuracy metrics. Results: RadiomiX achieved superior performance across four datasets: LLN (AUC = 0.850 and accuracy = 0.785), SLN (AUC = 0.845 and accuracy = 0.754), MBC (AUC = 0.889 and accuracy = 0.833), and CHE (AUC = 0.837 and accuracy = 0.730), significantly outperforming original published models (p < 0.001 for LLN/SLN and p = 0.023 for MBC accuracy). When original published models were re-evaluated using ten-fold cross-validation, their performance decreased substantially: LLN (AUC = 0.783 and accuracy = 0.731), SLN (AUC = 0.748 and accuracy = 0.714), MBC (AUC = 0.764 and accuracy = 0.711), and CHE (AUC = 0.755 and accuracy = 0.677), further highlighting RadiomiX’s methodological advantages. Conclusions: Systematically testing model combinations using RadiomiX has led to significant improvements in performance. This emphasizes the potential of automated ML as a step towards better-performing and more reliable radiomic models. Full article
(This article belongs to the Section Machine Learning and Artificial Intelligence in Diagnostics)
Show Figures

Figure 1

33 pages, 5542 KiB  
Review
Recent Advances in PET and Radioligand Therapy for Lung Cancer: FDG and FAP
by Eun Jeong Lee, Hyun Woo Chung, Young So, In Ae Kim, Hee Joung Kim and Kye Young Lee
Cancers 2025, 17(15), 2549; https://doi.org/10.3390/cancers17152549 - 1 Aug 2025
Viewed by 110
Abstract
Lung cancer is one of the most common cancers and the leading cause of cancer-related death worldwide. Despite advancements, the overall survival rate for lung cancer remains between 10% and 20% in most countries. However, recent progress in diagnostic tools and therapeutic strategies [...] Read more.
Lung cancer is one of the most common cancers and the leading cause of cancer-related death worldwide. Despite advancements, the overall survival rate for lung cancer remains between 10% and 20% in most countries. However, recent progress in diagnostic tools and therapeutic strategies has led to meaningful improvements in survival outcomes, highlighting the growing importance of personalized management based on accurate disease assessment. 18F-fluorodeoxyglucose positron emission tomography/computed tomography (FDG PET/CT) has become essential in the management of lung cancer, serving as a key imaging modality for initial diagnosis, staging, treatment response assessment, and follow-up evaluation. Recent developments in radiomics and artificial intelligence (AI), including machine learning and deep learning, have revolutionized the analysis of complex imaging data, enhancing the diagnostic and predictive capabilities of FDG PET/CT in lung cancer. However, the limitations of FDG, including its low specificity for malignancy, have driven the development of novel oncologic radiotracers. One such target is fibroblast activation protein (FAP), a type II transmembrane glycoprotein that is overexpressed in activated cancer-associated fibroblasts within the tumor microenvironment of various epithelial cancers. As a result, FAP-targeted radiopharmaceuticals represent a novel theranostic approach, offering the potential to integrate PET imaging with radioligand therapy (RLT). In this review, we provide a comprehensive overview of FDG PET/CT in lung cancer, along with recent advances in AI. Additionally, we discuss FAP-targeted radiopharmaceuticals for PET imaging and their potential application in RLT for the personalized management of lung cancer. Full article
(This article belongs to the Special Issue Molecular PET Imaging in Cancer Metabolic Studies)
Show Figures

Figure 1

35 pages, 887 KiB  
Review
Prognostic Factors in Colorectal Liver Metastases: An Exhaustive Review of the Literature and Future Prospectives
by Maria Conticchio, Emilie Uldry, Martin Hübner, Antonia Digklia, Montserrat Fraga, Christine Sempoux, Jean Louis Raisaro and David Fuks
Cancers 2025, 17(15), 2539; https://doi.org/10.3390/cancers17152539 - 31 Jul 2025
Viewed by 192
Abstract
Background: Colorectal liver metastasis (CRLM) represents a major clinical challenge in oncology, affecting 25–50% of colorectal cancer patients and significantly impacting survival. While multimodal therapies—including surgical resection, systemic chemotherapy, and local ablative techniques—have improved outcomes, prognosis remains heterogeneous due to variations in [...] Read more.
Background: Colorectal liver metastasis (CRLM) represents a major clinical challenge in oncology, affecting 25–50% of colorectal cancer patients and significantly impacting survival. While multimodal therapies—including surgical resection, systemic chemotherapy, and local ablative techniques—have improved outcomes, prognosis remains heterogeneous due to variations in tumor biology, patient factors, and institutional practices. Methods: This review synthesizes current evidence on prognostic factors influencing CRLM management, encompassing clinical (e.g., tumor burden, anatomic distribution, timing of metastases), biological (e.g., CEA levels, inflammatory markers), and molecular (e.g., RAS/BRAF mutations, MSI status, HER2 alterations) determinants. Results: Key findings highlight the critical role of molecular profiling in guiding therapeutic decisions, with RAS/BRAF mutations predicting resistance to anti-EGFR therapies and MSI-H status indicating potential responsiveness to immunotherapy. Emerging tools like circulating tumor DNA (ctDNA) and radiomics offer promise for dynamic risk stratification and early recurrence detection, while the gut microbiome is increasingly recognized as a modulator of treatment response. Conclusions: Despite advancements, challenges persist in standardizing resectability criteria and integrating multidisciplinary approaches. Current guidelines (NCCN, ESMO, ASCO) emphasize personalized strategies but lack granularity in terms of incorporating novel biomarkers. This exhaustive review underscores the imperative for the development of a unified, biomarker-integrated framework to refine CRLM management and improve long-term outcomes. Full article
Show Figures

Figure 1

14 pages, 2727 KiB  
Article
A Multimodal MRI-Based Model for Colorectal Liver Metastasis Prediction: Integrating Radiomics, Deep Learning, and Clinical Features with SHAP Interpretation
by Xin Yan, Furui Duan, Lu Chen, Runhong Wang, Kexin Li, Qiao Sun and Kuang Fu
Curr. Oncol. 2025, 32(8), 431; https://doi.org/10.3390/curroncol32080431 - 30 Jul 2025
Viewed by 182
Abstract
Purpose: Predicting colorectal cancer liver metastasis (CRLM) is essential for prognostic assessment. This study aims to develop and validate an interpretable multimodal machine learning framework based on multiparametric MRI for predicting CRLM, and to enhance the clinical interpretability of the model through [...] Read more.
Purpose: Predicting colorectal cancer liver metastasis (CRLM) is essential for prognostic assessment. This study aims to develop and validate an interpretable multimodal machine learning framework based on multiparametric MRI for predicting CRLM, and to enhance the clinical interpretability of the model through SHapley Additive exPlanations (SHAP) analysis and deep learning visualization. Methods: This multicenter retrospective study included 463 patients with pathologically confirmed colorectal cancer from two institutions, divided into training (n = 256), internal testing (n = 111), and external validation (n = 96) sets. Radiomics features were extracted from manually segmented regions on axial T2-weighted imaging (T2WI) and diffusion-weighted imaging (DWI). Deep learning features were obtained from a pretrained ResNet101 network using the same MRI inputs. A least absolute shrinkage and selection operator (LASSO) logistic regression classifier was developed for clinical, radiomics, deep learning, and combined models. Model performance was evaluated by AUC, sensitivity, specificity, and F1-score. SHAP was used to assess feature contributions, and Grad-CAM was applied to visualize deep feature attention. Results: The combined model integrating features across the three modalities achieved the highest performance across all datasets, with AUCs of 0.889 (training), 0.838 (internal test), and 0.822 (external validation), outperforming single-modality models. Decision curve analysis (DCA) revealed enhanced clinical net benefit from the integrated model, while calibration curves confirmed its good predictive consistency. SHAP analysis revealed that radiomic features related to T2WI texture (e.g., LargeDependenceLowGrayLevelEmphasis) and clinical biomarkers (e.g., CA19-9) were among the most predictive for CRLM. Grad-CAM visualizations confirmed that the deep learning model focused on tumor regions consistent with radiological interpretation. Conclusions: This study presents a robust and interpretable multiparametric MRI-based model for noninvasively predicting liver metastasis in colorectal cancer patients. By integrating handcrafted radiomics and deep learning features, and enhancing transparency through SHAP and Grad-CAM, the model provides both high predictive performance and clinically meaningful explanations. These findings highlight its potential value as a decision-support tool for individualized risk assessment and treatment planning in the management of colorectal cancer. Full article
(This article belongs to the Section Gastrointestinal Oncology)
Show Figures

Graphical abstract

23 pages, 4210 KiB  
Article
CT-Based Habitat Radiomics Combining Multi-Instance Learning for Early Prediction of Post-Neoadjuvant Lymph Node Metastasis in Esophageal Squamous Cell Carcinoma
by Qinghe Peng, Shumin Zhou, Runzhe Chen, Jinghui Pan, Xin Yang, Jinlong Du, Hongdong Liu, Hao Jiang, Xiaoyan Huang, Haojiang Li and Li Chen
Bioengineering 2025, 12(8), 813; https://doi.org/10.3390/bioengineering12080813 - 28 Jul 2025
Viewed by 377
Abstract
Early prediction of lymph node metastasis (LNM) following neoadjuvant therapy (NAT) is crucial for timely treatment optimization in esophageal squamous cell carcinoma (ESCC). This study developed and validated a computed tomography-based radiomic model for predicting pathologically confirmed LNM status at the time of [...] Read more.
Early prediction of lymph node metastasis (LNM) following neoadjuvant therapy (NAT) is crucial for timely treatment optimization in esophageal squamous cell carcinoma (ESCC). This study developed and validated a computed tomography-based radiomic model for predicting pathologically confirmed LNM status at the time of surgery in ESCC patients after NAT. A total of 469 ESCC patients from Sun Yat-sen University Cancer Center were retrospectively enrolled and randomized into a training cohort (n = 328) and a test cohort (n = 141). Three signatures were constructed: the tumor-habitat-based signature (Habitat_Rad), derived from radiomic features of three tumor subregions identified via K-means clustering; the multiple instance learning-based signature (MIL_Rad), combining features from 2.5D deep learning models; and the clinicoradiological signature (Clinic), developed through multivariate logistic regression. A combined radiomic nomogram integrating these signatures outperformed the individual models, achieving areas under the curve (AUCs) of 0.929 (95% CI, 0.901–0.957) and 0.852 (95% CI, 0.778–0.925) in the training and test cohorts, respectively. The decision curve analysis confirmed a high net clinical benefit, highlighting the nomogram’s potential for accurate LNM prediction after NAT and guiding individualized therapy. Full article
(This article belongs to the Special Issue Machine Learning Methods for Biomedical Imaging)
Show Figures

Graphical abstract

16 pages, 589 KiB  
Article
CT-Based Radiomics Enhance Respiratory Function Analysis for Lung SBRT
by Alice Porazzi, Mattia Zaffaroni, Vanessa Eleonora Pierini, Maria Giulia Vincini, Aurora Gaeta, Sara Raimondi, Lucrezia Berton, Lars Johannes Isaksson, Federico Mastroleo, Sara Gandini, Monica Casiraghi, Gaia Piperno, Lorenzo Spaggiari, Juliana Guarize, Stefano Maria Donghi, Łukasz Kuncman, Roberto Orecchia, Stefania Volpe and Barbara Alicja Jereczek-Fossa
Bioengineering 2025, 12(8), 800; https://doi.org/10.3390/bioengineering12080800 - 25 Jul 2025
Viewed by 452
Abstract
Introduction: Radiomics is the extraction of non-invasive and reproducible quantitative imaging features, which may yield mineable information for clinical practice implementation. Quantification of lung function through radiomics could play a role in the management of patients with pulmonary lesions. The aim of this [...] Read more.
Introduction: Radiomics is the extraction of non-invasive and reproducible quantitative imaging features, which may yield mineable information for clinical practice implementation. Quantification of lung function through radiomics could play a role in the management of patients with pulmonary lesions. The aim of this study is to test the capability of radiomic features to predict pulmonary function parameters, focusing on the diffusing capacity of lungs to carbon monoxide (DLCO). Methods: Retrospective data were retrieved from electronical medical records of patients treated with Stereotactic Body Radiation Therapy (SBRT) at a single institution. Inclusion criteria were as follows: (1) SBRT treatment performed for primary early-stage non-small cell lung cancer (ES-NSCLC) or oligometastatic lung nodules, (2) availability of simulation four-dimensional computed tomography (4DCT) scan, (3) baseline spirometry data availability, (4) availability of baseline clinical data, and (5) written informed consent for the anonymized use of data. The gross tumor volume (GTV) was segmented on 4DCT reconstructed phases representing the moment of maximum inhalation and maximum exhalation (Phase 0 and Phase 50, respectively), and radiomic features were extracted from the lung parenchyma subtracting the lesion/s. An iterative algorithm was clustered based on correlation, while keeping only those most associated with baseline and post-treatment DLCO. Three models were built to predict DLCO abnormality: the clinical model—containing clinical information; the radiomic model—containing the radiomic score; the clinical-radiomic model—containing clinical information and the radiomic score. For the models just described, the following were constructed: Model 1 based on the features in Phase 0; Model 2 based on the features in Phase 50; Model 3 based on the difference between the two phases. The AUC was used to compare their performances. Results: A total of 98 patients met the inclusion criteria. The Charlson Comorbidity Index (CCI) scored as the clinical variable most associated with baseline DLCO (p = 0.014), while the most associated features were mainly texture features and similar among the two phases. Clinical-radiomic models were the best at predicting both baseline and post-treatment abnormal DLCO. In particular, the performances for the three clinical-radiomic models at predicting baseline abnormal DLCO were AUC1 = 0.72, AUC2 = 0.72, and AUC3 = 0.75, for Model 1, Model 2, and Model 3, respectively. Regarding the prediction of post-treatment abnormal DLCO, the performances of the three clinical-radiomic models were AUC1 = 0.91, AUC2 = 0.91, and AUC3 = 0.95, for Model 1, Model 2, and Model 3, respectively. Conclusions: This study demonstrates that radiomic features extracted from healthy lung parenchyma on a 4DCT scan are associated with baseline pulmonary function parameters, showing that radiomics can add a layer of information in surrogate models for lung function assessment. Preliminary results suggest the potential applicability of these models for predicting post-SBRT lung function, warranting validation in larger, prospective cohorts. Full article
(This article belongs to the Special Issue Engineering the Future of Radiotherapy: Innovations and Challenges)
Show Figures

Figure 1

27 pages, 2617 KiB  
Article
Monte Carlo Gradient Boosted Trees for Cancer Staging: A Machine Learning Approach
by Audrey Eley, Thu Thu Hlaing, Daniel Breininger, Zarindokht Helforoush and Nezamoddin N. Kachouie
Cancers 2025, 17(15), 2452; https://doi.org/10.3390/cancers17152452 - 24 Jul 2025
Viewed by 334
Abstract
Machine learning algorithms are commonly employed for classification and interpretation of high-dimensional data. The classification task is often broken down into two separate procedures, and different methods are applied to achieve accurate results and produce interpretable outcomes. First, an effective subset of high-dimensional [...] Read more.
Machine learning algorithms are commonly employed for classification and interpretation of high-dimensional data. The classification task is often broken down into two separate procedures, and different methods are applied to achieve accurate results and produce interpretable outcomes. First, an effective subset of high-dimensional features must be extracted and then the selected subset will be used to train a classifier. Gradient Boosted Trees (GBT) is an ensemble model and, particularly due to their robustness, ability to model complex nonlinear interactions, and feature interpretability, they are well suited for complex applications. XGBoost (eXtreme Gradient Boosting) is a high-performance implementation of GBT that incorporates regularization, parallel computation, and efficient tree pruning that makes it a suitable efficient, interpretable, and scalable classifier with potential applications to medical data analysis. In this study, a Monte Carlo Gradient Boosted Trees (MCGBT) model is proposed for both feature reduction and classification. The proposed MCGBT method was applied to a lung cancer dataset for feature identification and classification. The dataset contains 107 radiomics which are quantitative imaging biomarkers extracted from CT scans. A reduced set of 12 radiomics were identified, and patients were classified into different cancer stages. Cancer staging accuracy of 90.3% across 100 independent runs was achieved which was on par with that obtained using the full set of 107 radiomics, enabling lean and deployable classifiers. Full article
(This article belongs to the Section Cancer Informatics and Big Data)
Show Figures

Figure 1

19 pages, 1282 KiB  
Article
The Role of Radiomic Analysis and Different Machine Learning Models in Prostate Cancer Diagnosis
by Eleni Bekou, Ioannis Seimenis, Athanasios Tsochatzis, Karafyllia Tziagkana, Nikolaos Kelekis, Savas Deftereos, Nikolaos Courcoutsakis, Michael I. Koukourakis and Efstratios Karavasilis
J. Imaging 2025, 11(8), 250; https://doi.org/10.3390/jimaging11080250 - 23 Jul 2025
Viewed by 331
Abstract
Prostate cancer (PCa) is the most common malignancy in men. Precise grading is crucial for the effective treatment approaches of PCa. Machine learning (ML) applied to biparametric Magnetic Resonance Imaging (bpMRI) radiomics holds promise for improving PCa diagnosis and prognosis. This study investigated [...] Read more.
Prostate cancer (PCa) is the most common malignancy in men. Precise grading is crucial for the effective treatment approaches of PCa. Machine learning (ML) applied to biparametric Magnetic Resonance Imaging (bpMRI) radiomics holds promise for improving PCa diagnosis and prognosis. This study investigated the efficiency of seven ML models to diagnose the different PCa grades, changing the input variables. Our studied sample comprised 214 men who underwent bpMRI in different imaging centers. Seven ML algorithms were compared using radiomic features extracted from T2-weighted (T2W) and diffusion-weighted (DWI) MRI, with and without the inclusion of Prostate-Specific Antigen (PSA) values. The performance of the models was evaluated using the receiver operating characteristic curve analysis. The models’ performance was strongly dependent on the input parameters. Radiomic features derived from T2WI and DWI, whether used independently or in combination, demonstrated limited clinical utility, with AUC values ranging from 0.703 to 0.807. However, incorporating the PSA index significantly improved the models’ efficiency, regardless of lesion location or degree of malignancy, resulting in AUC values ranging from 0.784 to 1.00. There is evidence that ML methods, in combination with radiomic analysis, can contribute to solving differential diagnostic problems of prostate cancers. Also, optimization of the analysis method is critical, according to the results of our study. Full article
(This article belongs to the Section Medical Imaging)
Show Figures

Figure 1

19 pages, 2931 KiB  
Article
Prediction of Breast Cancer Response to Neoadjuvant Therapy with Machine Learning: A Clinical, MRI-Qualitative, and Radiomics Approach
by Rami Hajri, Charles Aboudaram, Nathalie Lassau, Tarek Assi, Leony Antoun, Joana Mourato Ribeiro, Magali Lacroix-Triki, Samy Ammari and Corinne Balleyguier
Life 2025, 15(8), 1165; https://doi.org/10.3390/life15081165 - 23 Jul 2025
Viewed by 382
Abstract
Background: Pathological complete response (pCR) serves as a prognostic surrogate endpoint for long-term clinical outcomes in breast cancer patients receiving neoadjuvant systemic therapy (NAST). This study aims to develop and evaluate machine learning-based biomarkers for predicting pCR and recurrence-free survival (RFS). Methods: This [...] Read more.
Background: Pathological complete response (pCR) serves as a prognostic surrogate endpoint for long-term clinical outcomes in breast cancer patients receiving neoadjuvant systemic therapy (NAST). This study aims to develop and evaluate machine learning-based biomarkers for predicting pCR and recurrence-free survival (RFS). Methods: This retrospective monocentric study included 235 women (mean age 46 ± 11 years) with non-metastatic breast cancer treated with NAST. We developed various machine learning models using clinical features (age, genetic mutations, TNM stage, hormonal receptor expression, HER2 status, and histological grade), along with morphological features (size, T2 signal, and surrounding edema) and radiomics data extracted from pre-treatment MRI. Patients were divided into training and test groups with different MRI models. A customized machine learning pipeline was implemented to handle these diverse data types, consisting of feature selection and classification components. Results: The models demonstrated superior prediction ability using radiomics features, with the best model achieving an AUC of 0.72. Subgroup analysis revealed optimal performance in triple-negative breast cancer (AUC of 0.80) and HER2-positive subgroups (AUC of 0.65). Conclusion: Machine learning models incorporating clinical, qualitative, and radiomics data from pre-treatment MRI can effectively predict pCR in breast cancer patients receiving NAST, particularly among triple-negative and HER2-positive breast cancer subgroups. Full article
(This article belongs to the Special Issue New Insights Into Artificial Intelligence in Medical Imaging)
Show Figures

Figure 1

19 pages, 2950 KiB  
Article
Nomogram Based on the Most Relevant Clinical, CT, and Radiomic Features, and a Machine Learning Model to Predict EGFR Mutation Status in Non-Small Cell Lung Cancer
by Anass Benfares, Abdelali yahya Mourabiti, Badreddine Alami, Sara Boukansa, Ikram Benomar, Nizar El Bouardi, Moulay Youssef Alaoui Lamrani, Hind El Fatimi, Bouchra Amara, Mounia Serraj, Mohammed Smahi, Abdeljabbar Cherkaoui, Mamoun Qjidaa, Ahmed Lakhssassi, Mohammed Ouazzani Jamil, Mustapha Maaroufi and Hassan Qjidaa
J. Respir. 2025, 5(3), 11; https://doi.org/10.3390/jor5030011 - 23 Jul 2025
Viewed by 314
Abstract
Background: This study aimed to develop a nomogram based on the most relevant clinical, CT, and radiomic features comprising 11 key signatures (2 clinical, 2 CT-based, and 7 radiomic) for the non-invasive prediction of the EGFR mutation status and to support the timely [...] Read more.
Background: This study aimed to develop a nomogram based on the most relevant clinical, CT, and radiomic features comprising 11 key signatures (2 clinical, 2 CT-based, and 7 radiomic) for the non-invasive prediction of the EGFR mutation status and to support the timely initiation of tyrosine kinase inhibitor (TKI) therapy in patients with non-small cell lung cancer (NSCLC) adenocarcinoma. Methods: Retrospective real-world data were collected from 521 patients with histologically confirmed NSCLC adenocarcinoma who underwent CT imaging and either surgical resection or pathological biopsy for EGFR mutation testing. Five Random Forest classification models were developed and trained on various datasets constructed by combining clinical, CT, and radiomic features extracted from CT image regions of interest (ROIs), with and without feature preselection. Results: The model trained exclusively on the most relevant clinical, CT, and radiomic features demonstrated superior predictive performance compared to the other models, with strong discrimination between EGFR-mutant and wild-type cases (AUC = 0.88; macro-average = 0.90; micro-average = 0.89; precision = 0.90; recall = 0.94; F1-score = 0.91; and accuracy = 0.87). Conclusions: A nomogram constructed using a Random Forest model trained solely on the most informative clinical, CT, and radiomic features outperformed alternative approaches in the non-invasive prediction of the EGFR mutation status, offering a promising decision-support tool for precision treatment planning in NSCLC. Full article
Show Figures

Figure 1

23 pages, 3725 KiB  
Systematic Review
The Value of MRI-Based Radiomics in Predicting the Pathological Nodal Status of Rectal Cancer: A Systematic Review and Meta-Analysis
by David Luengo Gómez, Marta García Cerezo, David López Cornejo, Ángela Salmerón Ruiz, Encarnación González-Flores, Consolación Melguizo Alonso, Antonio Jesús Láinez Ramos-Bossini, José Prados and Francisco Gabriel Ortega Sánchez
Bioengineering 2025, 12(7), 786; https://doi.org/10.3390/bioengineering12070786 - 21 Jul 2025
Viewed by 349
Abstract
Background: MRI-based radiomics has emerged as a promising approach to enhance the non-invasive, presurgical assessment of lymph node staging in rectal cancer (RC). However, its clinical implementation remains limited due to methodological variability in published studies. We conducted a systematic review and meta-analysis [...] Read more.
Background: MRI-based radiomics has emerged as a promising approach to enhance the non-invasive, presurgical assessment of lymph node staging in rectal cancer (RC). However, its clinical implementation remains limited due to methodological variability in published studies. We conducted a systematic review and meta-analysis to synthesize the diagnostic performance of MRI-based radiomics models for predicting pathological nodal status (pN) in RC. Methods: A systematic literature search was conducted in PubMed, Web of Science, and Scopus for studies published until 31 December 2024. Eligible studies applied MRI-based radiomics for pN prediction in RC patients. We excluded other imaging sources and models combining radiomics and other data (e.g., clinical). All models with available outcome metrics were included in data analysis. Data extraction and quality assessment (QUADAS-2) were performed independently by two reviewers. Random-effects meta-analyses including hierarchical summary receiver operating characteristic (HSROC) and restricted maximum likelihood estimator (REML) analyses were conducted to pool sensitivity, specificity, area under the curve (AUC), and diagnostic odds ratios (DORs). Sensitivity analyses and publication bias evaluation were also performed. Results: Sixteen studies (n = 3157 patients) were included. The HSROC showed pooled sensitivity, specificity, and AUC values of 0.68 (95% CI, 0.63–0.72), 0.73 (95% CI, 0.68–0.78), and 0.70 (95% CI, 0.65–0.75), respectively. The mean pooled AUC and DOR obtained by REML were 0.78 (95% CI, 0.75–0.80) and 6.03 (95% CI, 4.65–7.82). Funnel plot asymmetry and Egger’s test (p = 0.025) indicated potential publication bias. Conclusions: Overall, MRI-based radiomics models demonstrated moderate accuracy in predicting pN status in RC, with some studies reporting outstanding results. However, heterogeneity in relevant methodological approaches such as the source of MRI sequences or machine learning methods applied along with possible publication bias call for further standardization and preclude their translation to clinical practice. Full article
Show Figures

Figure 1

15 pages, 2610 KiB  
Article
CT-Based Radiomics for a priori Predicting Response to Chemoradiation in Locally Advanced Lung Adenocarcinoma
by Erika Z. Chung, Laurentius O. Osapoetra, Patrick Cheung, Ian Poon, Alexander V. Louie, May Tsao, Yee Ung, Mateus T. Cunha, Ines B. Menjak and Gregory J. Czarnota
Cancers 2025, 17(14), 2386; https://doi.org/10.3390/cancers17142386 - 18 Jul 2025
Viewed by 306
Abstract
The standard treatment for patients with locally advanced non-small cell lung cancer (NSCLC) is concurrent chemoradiation. However, clinical responses are heterogeneous and generally not known until after the completion of therapy. Multiple studies have investigated imaging predictors (radiomics) for different cancer histologies, but [...] Read more.
The standard treatment for patients with locally advanced non-small cell lung cancer (NSCLC) is concurrent chemoradiation. However, clinical responses are heterogeneous and generally not known until after the completion of therapy. Multiple studies have investigated imaging predictors (radiomics) for different cancer histologies, but little exists for NSCLC. The objective of this study was to develop a multivariate CT-based radiomics model to a priori predict responses to definitive chemoradiation in patients with lung adenocarcinoma. Methods: Patients diagnosed with locally advanced unresectable lung adenocarcinoma who had undergone chemoradiotherapy followed by at least one dose of maintenance durvalumab were included. The PyRadiomics Python library was used to determine statistical, morphological, and textural features from normalized patient pre-treatment CT images and their wavelet-filtered versions. A nested leave-one-out cross-validation was used for model building and evaluation. Results: Fifty-seven patients formed the study cohort. The clinical stage was IIIA-C in 98% of patients. All but one received 6000–6600 cGy of radiation in 30–33 fractions. All received concurrent platinum-based chemotherapy. Based on RECIST 1.1, 20 (35%) patients were classified as responders (R) to chemoradiation and 37 (65%) patients as non-responders (NR). A three-feature model based on a KNN k = 1 machine learning classifier was found to have the best performance, achieving a recall, specificity, accuracy, balanced accuracy, precision, negative predictive value, F1-score, and area under the curve of 84%, 70%, 80%, 77%, 84%, 70%, 84%, and 0.77, respectively. Conclusions: Our results suggest that a CT-based radiomics model may be able to predict chemoradiation response for lung adenocarcinoma patients with estimated accuracies of 77–84%. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Figure 1

15 pages, 3326 KiB  
Article
Radiomics and Machine Learning Approaches for the Preoperative Classification of In Situ vs. Invasive Breast Cancer Using Dynamic Contrast-Enhanced Magnetic Resonance Imaging (DCE–MRI)
by Luana Conte, Rocco Rizzo, Alessandra Sallustio, Eleonora Maggiulli, Mariangela Capodieci, Francesco Tramacere, Alessandra Castelluccia, Giuseppe Raso, Ugo De Giorgi, Raffaella Massafra, Maurizio Portaluri, Donato Cascio and Giorgio De Nunzio
Appl. Sci. 2025, 15(14), 7999; https://doi.org/10.3390/app15147999 - 18 Jul 2025
Viewed by 323
Abstract
Accurate preoperative distinction between in situ and invasive Breast Cancer (BC) is critical for clinical decision-making and treatment planning. Radiomics and Machine Learning (ML) have shown promise in enhancing diagnostic performance from breast MRI, yet their application to this specific task remains underexplored. [...] Read more.
Accurate preoperative distinction between in situ and invasive Breast Cancer (BC) is critical for clinical decision-making and treatment planning. Radiomics and Machine Learning (ML) have shown promise in enhancing diagnostic performance from breast MRI, yet their application to this specific task remains underexplored. The aim of this study was to evaluate the performance of several ML classifiers, trained on radiomic features extracted from DCE–MRI and supported by basic clinical information, for the classification of in situ versus invasive BC lesions. In this study, we retrospectively analysed 71 post-contrast DCE–MRI scans (24 in situ, 47 invasive cases). Radiomic features were extracted from manually segmented tumour regions using the PyRadiomics library, and a limited set of basic clinical variables was also included. Several ML classifiers were evaluated in a Leave-One-Out Cross-Validation (LOOCV) scheme. Feature selection was performed using two different strategies: Minimum Redundancy Maximum Relevance (MRMR), mutual information. Axial 3D rotation was used for data augmentation. Support Vector Machine (SVM), K Nearest Neighbors (KNN), Random Forest (RF), and Extreme Gradient Boosting (XGBoost) were the best-performing models, with an Area Under the Curve (AUC) ranging from 0.77 to 0.81. Notably, KNN achieved the best balance between sensitivity and specificity without the need for data augmentation. Our findings confirm that radiomic features extracted from DCE–MRI, combined with well-validated ML models, can effectively support the differentiation of in situ vs. invasive breast cancer. This approach is quite robust even in small datasets and may aid in improving preoperative planning. Further validation on larger cohorts and integration with additional imaging or clinical data are recommended. Full article
Show Figures

Figure 1

22 pages, 368 KiB  
Review
Early Detection of Pancreatic Cancer: Current Advances and Future Opportunities
by Zijin Lin, Esther A. Adeniran, Yanna Cai, Touseef Ahmad Qureshi, Debiao Li, Jun Gong, Jianing Li, Stephen J. Pandol and Yi Jiang
Biomedicines 2025, 13(7), 1733; https://doi.org/10.3390/biomedicines13071733 - 15 Jul 2025
Viewed by 711
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains among the most lethal malignancies, with a five-year survival rate below 12%, largely attributable to its asymptomatic onset, late-stage diagnosis, and limited curative treatment options. Although PDAC accounts for approximately 3% of all cancers, it is projected to [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) remains among the most lethal malignancies, with a five-year survival rate below 12%, largely attributable to its asymptomatic onset, late-stage diagnosis, and limited curative treatment options. Although PDAC accounts for approximately 3% of all cancers, it is projected to become the second leading cause of cancer-related mortality in the United States by 2030. A major contributor to its dismal prognosis is the lack of validated early detection strategies for asymptomatic individuals. In this review, we present a comprehensive synthesis of current advances in the early detection of PDAC, with a focus on the identification of high-risk populations, novel biomarker platforms, advanced imaging modalities, and artificial intelligence (AI)-driven tools. We highlight high-risk groups—such as those with new-onset diabetes after age 50, pancreatic steatosis, chronic pancreatitis, cystic precursor lesions, and hereditary cancer syndromes—as priority populations for targeted surveillance. Novel biomarker panels, including circulating tumor DNA (ctDNA), miRNAs, and exosomes, have demonstrated improved diagnostic accuracy in early-stage disease. Recent developments in imaging, such as multiparametric MRI, contrast-enhanced endoscopic ultrasound, and molecular imaging, offer improved sensitivity in detecting small or precursor lesions. AI-enhanced radiomics and machine learning models applied to prediagnostic CT scans and electronic health records are emerging as valuable tools for risk prediction prior to clinical presentation. We further refine the Define–Enrich–Find (DEF) framework to propose a clinically actionable strategy that integrates these innovations. Collectively, these advances pave the way for personalized, multimodal surveillance strategies with the potential to improve outcomes in this historically challenging malignancy. Full article
Show Figures

Graphical abstract

Back to TopTop