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

Search Results (117)

Search Parameters:
Keywords = multiparametric radiomics

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
27 pages, 1211 KB  
Review
Locally Advanced Cervical Cancer: Multiparametric MRI in Gynecologic Oncology and Precision Medicine
by Sara Boemi, Matilde Pavan, Roberta Siena, Carla Lo Giudice, Alessia Pagana, Marco Marzio Panella and Maria Teresa Bruno
Diagnostics 2025, 15(22), 2858; https://doi.org/10.3390/diagnostics15222858 - 12 Nov 2025
Viewed by 48
Abstract
Background: Locally advanced cervical cancer (LACC) represents a significant challenge in oncology, requiring accurate assessment of local extent and metastatic spread. Multiparametric magnetic resonance imaging (mpMRI) has assumed a central role in the loco-regional characterization of the tumor due to its high soft-tissue [...] Read more.
Background: Locally advanced cervical cancer (LACC) represents a significant challenge in oncology, requiring accurate assessment of local extent and metastatic spread. Multiparametric magnetic resonance imaging (mpMRI) has assumed a central role in the loco-regional characterization of the tumor due to its high soft-tissue resolution and the ability to integrate functional information. Objectives: In this narrative review, we explore the use of mpMRI in the diagnosis, staging, and treatment response of LACC, comparing its performance with that of PET/CT, which remains complementary for remote staging. The potential of whole-body magnetic resonance imaging (WB-MRI) and hybrid PET/MRI techniques is also analyzed, as well as the emerging applications of radiomics and artificial intelligence. The paper also discusses technical limitations, interpretative variability, and the importance of protocol standardization. The goal is to provide an updated and translational summary of imaging in LACC, with implications for clinical practice and future research. Methods: Prospective and retrospective studies, systematic reviews, and meta-analyses on adult patients with cervical cancer were included. Results: Fifty-two studies were included. MRI demonstrated a sensitivity and specificity greater than 80% for parametrial and bladder invasion, but limited sensitivity (45–60%) for lymph node disease, lower than PET/CT. Multiparametric MRI was useful in early prediction of response to chemotherapy and radiotherapy and in distinguishing residual disease from fibrosis. The integration of MRI into Image-Guided Adaptive Brachytherapy (IGABT) resulted in improved oncological outcomes and reduced toxicity. The applications of radiomics and AI demonstrated enormous potential in predicting therapeutic response and lymph node status in the MRI study, but multicenter validation is still needed. Conclusions: MRI is the cornerstone of the local–regional staging of advanced cervical cancer; it has become an essential and crucial tool in treatment planning. Its use, combined with PET/CT for lymph node assessment and metastatic disease staging, is now the standard of care. Future prospects include the use of whole-body MRI and the development of predictive models based on radiomics and artificial intelligence. Full article
(This article belongs to the Section Medical Imaging and Theranostics)
Show Figures

Figure 1

21 pages, 3119 KB  
Review
Next-Generation Advances in Prostate Cancer Imaging and Artificial Intelligence Applications
by Kathleen H. Miao, Julia H. Miao, Mark Finkelstein, Aritrick Chatterjee and Aytekin Oto
J. Imaging 2025, 11(11), 390; https://doi.org/10.3390/jimaging11110390 - 3 Nov 2025
Viewed by 526
Abstract
Prostate cancer is one of the leading causes of cancer-related morbidity and mortality worldwide, and imaging plays a critical role in its detection, localization, staging, treatment, and management. The advent of artificial intelligence (AI) has introduced transformative possibilities in prostate imaging, offering enhanced [...] Read more.
Prostate cancer is one of the leading causes of cancer-related morbidity and mortality worldwide, and imaging plays a critical role in its detection, localization, staging, treatment, and management. The advent of artificial intelligence (AI) has introduced transformative possibilities in prostate imaging, offering enhanced accuracy, efficiency, and consistency. This review explores the integration of AI in prostate cancer diagnostics across key imaging modalities, including multiparametric MRI (mpMRI), PSMA PET/CT, and transrectal ultrasound (TRUS). Advanced AI technologies, such as machine learning, deep learning, and radiomics, are being applied for lesion detection, risk stratification, segmentation, biopsy targeting, and treatment planning. AI-augmented systems have demonstrated the ability to support PI-RADS scoring, automate prostate and tumor segmentation, guide targeted biopsies, and optimize radiation therapy. Despite promising performance, challenges persist regarding data heterogeneity, algorithm generalizability, ethical considerations, and clinical implementation. Looking ahead, multimodal AI models integrating imaging, genomics, and clinical data hold promise for advancing precision medicine in prostate cancer care and assisting clinicians, particularly in underserved regions with limited access to specialists. Continued multidisciplinary collaboration will be essential to translate these innovations into evidence-based practice. This article explores current AI applications and future directions that are transforming prostate imaging and patient care. Full article
(This article belongs to the Special Issue Celebrating the 10th Anniversary of the Journal of Imaging)
Show Figures

Figure 1

20 pages, 3745 KB  
Article
Using Delta MRI-Based Radiomics for Monitoring Early Peri-Tumoral Changes in a Mouse Model of Glioblastoma: Primary Study
by Haitham Al-Mubarak and Mohammed S. Alshuhri
Cancers 2025, 17(21), 3545; https://doi.org/10.3390/cancers17213545 - 1 Nov 2025
Viewed by 345
Abstract
Background/Objectives: Glioblastoma (GBM) is an aggressive primary brain tumor marked by diffuse infiltration into surrounding brain tissue. The peritumoral zone often appears normal on imaging yet harbors microscopic invasion. While perfusion-based studies, such as arterial spin labeling (ASL), have profiled this region, longitudinal [...] Read more.
Background/Objectives: Glioblastoma (GBM) is an aggressive primary brain tumor marked by diffuse infiltration into surrounding brain tissue. The peritumoral zone often appears normal on imaging yet harbors microscopic invasion. While perfusion-based studies, such as arterial spin labeling (ASL), have profiled this region, longitudinal radiomic monitoring remains limited. This study investigates delta radiomics using multiparametric MRI (mpMRI) in a GBM mouse model to track subtle peritumoral changes over time. Methods: A G7 GBM xenograft model was established in nine nude mice, imaged at 9- and 12 weeks post-implantation using MRI (T1W, T2W, T2 mapping, DWI-ADC, FA, and ASL) and co-registered histopathology (H&E, HLA staining). Tumor and peritumoral regions were manually segmented, and 107 radiomic features (shape, first-order, texture) were extracted per sequence and histology. The delta features were calculated and compared between timepoints. Results: The robust T2W texture and T2 map first-order features demonstrated the greatest sensitivity and reproducibility in capturing temporal peritumoral brain zone changes, distinguishing between time points used by K-mean. Conclusions: Delta radiomics offers added value over static analysis for early monitoring of peritumoral brain zone changes. The first-order and texture features of radiomics could serve as robust biomarkers of peritumoral invasion. These findings highlight the potential of longitudinal MRI-based radiomics to characterize glioblastoma progression and inform translational research. Full article
(This article belongs to the Section Methods and Technologies Development)
Show Figures

Figure 1

26 pages, 1421 KB  
Systematic Review
Improving Early Prostate Cancer Detection Through Artificial Intelligence: Evidence from a Systematic Review
by Vincenzo Ciccone, Marina Garofano, Rosaria Del Sorbo, Gabriele Mongelli, Mariella Izzo, Francesco Negri, Roberta Buonocore, Francesca Salerno, Rosario Gnazzo, Gaetano Ungaro and Alessia Bramanti
Cancers 2025, 17(21), 3503; https://doi.org/10.3390/cancers17213503 - 30 Oct 2025
Viewed by 343
Abstract
Background: Prostate cancer is one of the most common malignancies in men and a leading cause of cancer-related mortality. Early detection is essential to ensure curative treatment and favorable outcomes, but traditional diagnostic approaches—such as serum prostate-specific antigen (PSA) testing, digital rectal examination [...] Read more.
Background: Prostate cancer is one of the most common malignancies in men and a leading cause of cancer-related mortality. Early detection is essential to ensure curative treatment and favorable outcomes, but traditional diagnostic approaches—such as serum prostate-specific antigen (PSA) testing, digital rectal examination (DRE), and histopathological confirmation following biopsy—are limited by suboptimal accuracy and variability. Multiparametric magnetic resonance imaging (mpMRI) has improved diagnostic performance but remains highly dependent on reader expertise. Artificial intelligence (AI) offers promising opportunities to enhance diagnostic accuracy, reproducibility, and efficiency in prostate cancer detection. Objective: To evaluate the diagnostic accuracy and reporting timeliness of AI-based technologies compared with conventional diagnostic methods in the early detection of prostate cancer. Methods: Following PRISMA 2020 guidelines, PubMed, Scopus, Web of Science, and Cochrane Library were searched for studies published between January 2015 and April 2025. Eligible designs included randomized controlled trials, cohort, case–control, and pilot studies applying AI-based technologies to early prostate cancer diagnosis. Data on AUC-ROC, sensitivity, specificity, predictive values, diagnostic odds ratio (DOR), and time-to-reporting were narratively synthesized due to heterogeneity. Risk of bias was assessed using the QUADAS-AI tool. Results: Twenty-three studies involving 23,270 patients were included. AI-based technologies achieved a median AUC-ROC of 0.88 (range 0.70–0.93), with median sensitivity and specificity of 0.86 and 0.83, respectively. Compared with radiologists, AI or AI-assisted readings improved or matched diagnostic accuracy, reduced inter-reader variability, and decreased reporting time by up to 56%. Conclusions: AI-based technologies show strong diagnostic performance in early prostate cancer detection. However, methodological heterogeneity and limited standardization restrict generalizability. Large-scale prospective trials are required to validate clinical integration. Full article
(This article belongs to the Special Issue Medical Imaging and Artificial Intelligence in Cancer)
Show Figures

Figure 1

13 pages, 881 KB  
Article
Radiomics and Deep Learning Interplay for Predicting MGMT Methylation in Glioblastoma: The Crucial Role of Segmentation Quality
by Francesca Lizzi, Sara Saponaro, Alessia Giuliano, Cinzia Talamonti, Leonardo Ubaldi and Alessandra Retico
Cancers 2025, 17(21), 3417; https://doi.org/10.3390/cancers17213417 - 24 Oct 2025
Viewed by 364
Abstract
Background/Objectives: Glioblastoma (GBM) is the most malignant subtype of glioma and shows the poorest prognosis with a median survival time of 15 months. The methylation status of the Methylguanine-DNA Methyltransferase (MGMT) was proven to be a crucial factor in selecting the most appropriate [...] Read more.
Background/Objectives: Glioblastoma (GBM) is the most malignant subtype of glioma and shows the poorest prognosis with a median survival time of 15 months. The methylation status of the Methylguanine-DNA Methyltransferase (MGMT) was proven to be a crucial factor in selecting the most appropriate therapy. Currently, it is assessed through brain biopsy, which is a highly invasive and very expensive technique. For these reasons, in recent years, the possibility of inferring this information from multi-parametric Magnetic Resonance Imaging (mpMRI) has been widely explored. However, substantial differences in performance are reported in the literature. Methods: In this study, we developed several models based on either radiomic or deep learning approaches and a mixture of them using mpMRI for the MGMT status assessment using the public dataset UPENN-GBM, available on The Cancer Imaging Archive. Despite the tests performed using all MRI acquisitions and different methodological approaches, we did not obtain sufficiently reliable performance to direct the therapeutic path of patients. We thus investigated the impact of segmentation quality on MGMT status prediction since the UPENN-GBM dataset contains both automatic and manual refined segmentation masks. Results: We found that performance obtained through radiomic features computed on manually segmented tumors was significantly higher compared to that obtained using automatic segmentation, even when the differences between segmentation masks, measured in terms of Dice Similarity Coefficient (DSC), is not significantly different. Conclusion: This could be the reason why very different MGMT classification performance is typically reported and suggests the creation of a benchmark dataset, with high-quality segmentation masks. Full article
(This article belongs to the Special Issue The Development and Application of Imaging Biomarkers in Cancer)
Show Figures

Figure 1

16 pages, 254 KB  
Review
Advanced Neuroimaging and Emerging Systemic Therapies in Glioblastoma: Current Evidence and Future Directions
by Ilona Bar-Letkiewicz, Anna Pieczyńska, Małgorzata Dudzic, Michał Szkudlarek, Krystyna Adamska and Katarzyna Hojan
Biomedicines 2025, 13(11), 2597; https://doi.org/10.3390/biomedicines13112597 - 23 Oct 2025
Viewed by 738
Abstract
Despite technological progress, glioblastoma (GBM) continues to confer dismal prognoses. Modern neuroimaging methods are assuming an ever greater role in diagnosing and monitoring brain tumors. This review shows current neuroimaging approaches and systemic therapeutic strategies for glioblastoma, with a focus on emerging and [...] Read more.
Despite technological progress, glioblastoma (GBM) continues to confer dismal prognoses. Modern neuroimaging methods are assuming an ever greater role in diagnosing and monitoring brain tumors. This review shows current neuroimaging approaches and systemic therapeutic strategies for glioblastoma, with a focus on emerging and innovative treatments. Advances in multiparametric magnetic resonance imaging—MRI (diffusion, perfusion, and spectroscopy) and novel positron emission tomography (PET) tracers, complemented by radiomics and artificial intelligence (AI), now refine tumor delineation, differentiate progression from treatment effects, and may help predict treatment responses. Maximal safe resection followed by chemoradiotherapy with temozolomide remains the standard, with the greatest benefit seen in O6-methylguanine DNA methyltransferase (MGMT) promoter-methylated tumors. Bevacizumab and other targeted modalities offer mainly progression-free, not overall survival, gains. Immune checkpoint inhibitors (e.g., nivolumab) have not improved survival in unselected GBM, while early multi-antigen CAR-T (chimeric antigen receptor T-cell) strategies show preliminary bioactivity without established durability. While actionable alterations (NTRK fusions and BRAF V600E) justify selective targeted therapy trials, their definitive benefit in classical GBM is unproven. Future priorities include harmonized imaging molecular integration, AI-driven prognostic modeling, novel PET tracers, and strategies to breach or transiently open the blood–brain barrier to enhance drug delivery. Convergence of these domains may convert diagnostic precision into improved patient outcomes. Full article
(This article belongs to the Special Issue Medical Imaging in Brain Tumor: Charting the Future)
24 pages, 1024 KB  
Review
Artificial Intelligence in Glioma Diagnosis: A Narrative Review of Radiomics and Deep Learning for Tumor Classification and Molecular Profiling Across Positron Emission Tomography and Magnetic Resonance Imaging
by Rafail C. Christodoulou, Rafael Pitsillos, Platon S. Papageorgiou, Vasileia Petrou, Georgios Vamvouras, Ludwing Rivera, Sokratis G. Papageorgiou, Elena E. Solomou and Michalis F. Georgiou
Eng 2025, 6(10), 262; https://doi.org/10.3390/eng6100262 - 3 Oct 2025
Viewed by 1465
Abstract
Background: This narrative review summarizes recent progress in artificial intelligence (AI), especially radiomics and deep learning, for non-invasive diagnosis and molecular profiling of gliomas. Methodology: A thorough literature search was conducted on PubMed, Scopus, and Embase for studies published from January [...] Read more.
Background: This narrative review summarizes recent progress in artificial intelligence (AI), especially radiomics and deep learning, for non-invasive diagnosis and molecular profiling of gliomas. Methodology: A thorough literature search was conducted on PubMed, Scopus, and Embase for studies published from January 2020 to July 2025, focusing on clinical and technical research. In key areas, these studies examine AI models’ predictive capabilities with multi-parametric Magnetic Resonance Imaging (MRI) and Positron Emission Tomography (PET). Results: The domains identified in the literature include the advancement of radiomic models for tumor grading and biomarker prediction, such as Isocitrate Dehydrogenase (IDH) mutation, O6-methylguanine-dna methyltransferase (MGMT) promoter methylation, and 1p/19q codeletion. The growing use of convolutional neural networks (CNNs) and generative adversarial networks (GANs) in tumor segmentation, classification, and prognosis was also a significant topic discussed in the literature. Deep learning (DL) methods are evaluated against traditional radiomics regarding feature extraction, scalability, and robustness to imaging protocol differences across institutions. Conclusions: This review analyzes emerging efforts to combine clinical, imaging, and histology data within hybrid or transformer-based AI systems to enhance diagnostic accuracy. Significant findings include the application of DL to predict cyclin-dependent kinase inhibitor 2A/B (CDKN2A/B) deletion and chemokine CCL2 expression. These highlight the expanding capabilities of imaging-based genomic inference and the importance of clinical data in multimodal fusion. Challenges such as data harmonization, model interpretability, and external validation still need to be addressed. Full article
Show Figures

Figure 1

15 pages, 2112 KB  
Article
Radiomics-Based Preoperative Assessment of Muscle-Invasive Bladder Cancer Using Combined T2 and ADC MRI: A Multicohort Validation Study
by Dmitry Kabanov, Natalia Rubtsova, Aleksandra Golbits, Andrey Kaprin, Valentin Sinitsyn and Mikhail Potievskiy
J. Imaging 2025, 11(10), 342; https://doi.org/10.3390/jimaging11100342 - 1 Oct 2025
Viewed by 563
Abstract
Accurate preoperative staging of bladder cancer on MRI remains challenging because visual reads vary across observers. We investigated a multiparametric MRI (mpMRI) radiomics approach to predict muscle invasion (≥T2) and prospectively tested it on a validation cohort. Eighty-four patients with urothelial carcinoma underwent [...] Read more.
Accurate preoperative staging of bladder cancer on MRI remains challenging because visual reads vary across observers. We investigated a multiparametric MRI (mpMRI) radiomics approach to predict muscle invasion (≥T2) and prospectively tested it on a validation cohort. Eighty-four patients with urothelial carcinoma underwent 1.5-T mpMRI per VI-RADS (T2-weighted imaging and DWI-derived ADC maps). Two blinded radiologists performed 3D tumor segmentation; 37 features per sequence were extracted (LifeX) using absolute resampling. In the training cohort (n = 40), features that differed between non-muscle-invasive and muscle-invasive tumors (Mann–Whitney p < 0.05) underwent ROC analysis with cut-offs defined by the Youden index. A compact descriptor combining GLRLM-LRLGE from T2 and GLRLM-SRLGE from ADC was then fixed and applied without re-selection to a prospective validation cohort (n = 44). Histopathology within 6 weeks—TURBT or cystectomy—served as the reference. Eleven T2-based and fifteen ADC-based features pointed to invasion; DWI texture features were not informative. The descriptor yielded AUCs of 0.934 (training) and 0.871 (validation) with 85.7% sensitivity and 96.2% specificity in validation. Collectively, these findings indicate that combined T2/ADC radiomics can provide high diagnostic accuracy and may serve as a useful decision support tool, after multicenter, multi-vendor validation. Full article
(This article belongs to the Topic Machine Learning and Deep Learning in Medical Imaging)
Show Figures

Figure 1

16 pages, 8673 KB  
Article
PHSP-Net: Personalized Habitat-Aware Deep Learning for Multi-Center Glioblastoma Survival Prediction Using Multiparametric MRI
by Tianci Liu, Yao Zheng, Chengwei Chen, Jie Wei, Dong Huang, Yuefei Feng and Yang Liu
Bioengineering 2025, 12(9), 978; https://doi.org/10.3390/bioengineering12090978 - 15 Sep 2025
Viewed by 701
Abstract
Background: Glioblastoma (GBM) is a highly aggressive and heterogeneous primary malignancy of the central nervous system, with a median overall survival (OS) of approximately 15 months. Achieving accurate and generalizable OS prediction across multi-center settings is essential for clinical application. Methods: We propose [...] Read more.
Background: Glioblastoma (GBM) is a highly aggressive and heterogeneous primary malignancy of the central nervous system, with a median overall survival (OS) of approximately 15 months. Achieving accurate and generalizable OS prediction across multi-center settings is essential for clinical application. Methods: We propose a Personalized Habitat-aware Survival Prediction Network (PHSP-Net) that integrates multiparametric MRI with an adaptive habitat partitioning strategy. The network combines deep convolutional feature extraction and interpretable visualization modules to perform patient-specific subregional segmentation and survival prediction. A total of 1084 patients with histologically confirmed WHO grade IV GBM from four centers (UPENN-GBM, UCSF-PDGM, LUMIERE and TCGA-GBM) were included. PHSP-Net was compared with conventional radiomics, habitat imaging models and ResNet10, with independent validation on two external cohorts. Results: PHSP-Net achieved an AUROC of 0.795 (95% CI: 0.731–0.852) in the internal validation set, and 0.707 and 0.726 in the LUMIERE and TCGA-GBM external test sets, respectively—outperforming both comparison models. Kaplan–Meier analysis revealed significant OS differences between predicted high- and low-risk groups (log-rank p < 0.05). Visualization analysis indicated that necrotic-region habitats were key prognostic indicators. Conclusions: PHSP-Net demonstrates high predictive accuracy, robust cross-center generalization and improved interpretability in multi-center GBM cohorts. By enabling personalized habitat visualization, it offers a promising non-invasive tool for prognostic assessment and individualized clinical decision making in GBM. Full article
(This article belongs to the Special Issue Modern Medical Imaging in Disease Diagnosis Applications)
Show Figures

Graphical abstract

21 pages, 2336 KB  
Article
Machine and Deep Learning on Radiomic Features from Contrast-Enhanced Mammography and Dynamic Contrast-Enhanced Magnetic Resonance Imaging for Breast Cancer Characterization
by Roberta Fusco, Vincenza Granata, Teresa Petrosino, Paolo Vallone, Maria Assunta Daniela Iasevoli, Mauro Mattace Raso, Sergio Venanzio Setola, Davide Pupo, Gerardo Ferrara, Annarita Fanizzi, Raffaella Massafra, Miria Lafranceschina, Daniele La Forgia, Laura Greco, Francesca Romana Ferranti, Valeria De Soccio, Antonello Vidiri, Francesca Botta, Valeria Dominelli, Enrico Cassano, Charlotte Marguerite Lucille Trombadori, Paolo Belli, Giovanna Trecate, Chiara Tenconi, Maria Carmen De Santis, Luca Boldrini and Antonella Petrilloadd Show full author list remove Hide full author list
Bioengineering 2025, 12(9), 952; https://doi.org/10.3390/bioengineering12090952 - 2 Sep 2025
Viewed by 1371
Abstract
Objective: The aim of this study was to evaluate the accuracy of machine and deep learning approaches on radiomics features obtained by Dynamic Contrast Enhanced Magnetic Resonance Imaging (DCE-MRI) and contrast enhanced mammography (CEM) in the characterization of breast cancer and in the [...] Read more.
Objective: The aim of this study was to evaluate the accuracy of machine and deep learning approaches on radiomics features obtained by Dynamic Contrast Enhanced Magnetic Resonance Imaging (DCE-MRI) and contrast enhanced mammography (CEM) in the characterization of breast cancer and in the prediction of the tumor molecular profile. Methods: A total of 153 patients with malignant and benign lesions were analyzed and underwent MRI examinations. Considering the histological findings as the ground truth, three different types of findings were used in the analysis: (1) benign versus malignant lesions; (2) G1 + G2 vs. G3 classification; (3) the presence of human epidermal growth factor receptor 2 (HER2+ vs. HER2−). Radiomic features (n = 851) were extracted from manually segmented regions of interest using the PyRadiomics platform, following IBSI-compliant protocols. Highly correlated features were excluded, and the remaining features were standardized using z-score normalization. A feature selection process based on Elastic Net regularization (α = 0.5) was used to reduce dimensionality. Synthetic balancing of the training data was applied using the ROSE method to address class imbalance. Model performance was evaluated using repeated 10-fold cross-validation and AUC-based metrics. Results: Among the 153 patients enrolled in the studies, 113 were malignant lesions. Among the 113 malignant lesions, 32 had high grading (G3) and 66 had the HER2+ receptor. Radiomic features derived from both CEM and DCE-MRI showed strong discriminative performance for malignancy detection, with several features achieving AUCs above 0.80. Gradient Boosting Machine (GBM) achieved the highest accuracy (0.911) and AUC (0.907) in differentiating benign from malignant lesions. For tumor grading, the neural network model attained the best accuracy (0.848), while LASSO yielded the highest sensitivity (0.667) for detecting high-grade tumors. In predicting HER2+ status, the neural network also performed best (AUC = 0.669), with a sensitivity of 0.842. Conclusions: Radiomics-based machine learning models applied to multiparametric CEM and DCE-MRI images offer promising, non-invasive tools for breast cancer characterization. The models effectively distinguished benign from malignant lesions and showed potential in predicting histological grade and HER2 status. These results demonstrate that radiomic features extracted from CEM and DCE-MRI, when analyzed through machine and deep learning models, can support accurate breast cancer characterization. Such models may assist clinicians in early diagnosis, histological grading, and biomarker assessment, potentially enhancing personalized treatment planning and non-invasive decision-making in routine practice. Full article
Show Figures

Figure 1

14 pages, 2727 KB  
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 1672
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

22 pages, 368 KB  
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
Cited by 1 | Viewed by 9129
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

28 pages, 1727 KB  
Review
Computational and Imaging Approaches for Precision Characterization of Bone, Cartilage, and Synovial Biomolecules
by Rahul Kumar, Kyle Sporn, Vibhav Prabhakar, Ahab Alnemri, Akshay Khanna, Phani Paladugu, Chirag Gowda, Louis Clarkson, Nasif Zaman and Alireza Tavakkoli
J. Pers. Med. 2025, 15(7), 298; https://doi.org/10.3390/jpm15070298 - 9 Jul 2025
Viewed by 1626
Abstract
Background/Objectives: Degenerative joint diseases (DJDs) involve intricate molecular disruptions within bone, cartilage, and synovial tissues, often preceding overt radiographic changes. These tissues exhibit complex biomolecular architectures and their degeneration leads to microstructural disorganization and inflammation that are challenging to detect with conventional imaging [...] Read more.
Background/Objectives: Degenerative joint diseases (DJDs) involve intricate molecular disruptions within bone, cartilage, and synovial tissues, often preceding overt radiographic changes. These tissues exhibit complex biomolecular architectures and their degeneration leads to microstructural disorganization and inflammation that are challenging to detect with conventional imaging techniques. This review aims to synthesize recent advances in imaging, computational modeling, and sequencing technologies that enable high-resolution, non-invasive characterization of joint tissue health. Methods: We examined advanced modalities including high-resolution MRI (e.g., T1ρ, sodium MRI), quantitative and dual-energy CT (qCT, DECT), and ultrasound elastography, integrating them with radiomics, deep learning, and multi-scale modeling approaches. We also evaluated RNA-seq, spatial transcriptomics, and mass spectrometry-based proteomics for omics-guided imaging biomarker discovery. Results: Emerging technologies now permit detailed visualization of proteoglycan content, collagen integrity, mineralization patterns, and inflammatory microenvironments. Computational frameworks ranging from convolutional neural networks to finite element and agent-based models enhance diagnostic granularity. Multi-omics integration links imaging phenotypes to gene and protein expression, enabling predictive modeling of tissue remodeling, risk stratification, and personalized therapy planning. Conclusions: The convergence of imaging, AI, and molecular profiling is transforming musculoskeletal diagnostics. These synergistic platforms enable early detection, multi-parametric tissue assessment, and targeted intervention. Widespread clinical integration requires robust data infrastructure, regulatory compliance, and physician education, but offers a pathway toward precision musculoskeletal care. Full article
(This article belongs to the Special Issue Cutting-Edge Diagnostics: The Impact of Imaging on Precision Medicine)
Show Figures

Figure 1

17 pages, 4423 KB  
Article
Multivariate Framework of Metabolism in Advanced Prostate Cancer Using Whole Abdominal and Pelvic Hyperpolarized 13C MRI—A Correlative Study with Clinical Outcomes
by Hsin-Yu Chen, Ivan de Kouchkovsky, Robert A. Bok, Michael A. Ohliger, Zhen J. Wang, Daniel Gebrezgiabhier, Tanner Nickles, Lucas Carvajal, Jeremy W. Gordon, Peder E. Z. Larson, John Kurhanewicz, Rahul Aggarwal and Daniel B. Vigneron
Cancers 2025, 17(13), 2211; https://doi.org/10.3390/cancers17132211 - 1 Jul 2025
Cited by 1 | Viewed by 1320 | Correction
Abstract
Background: Most of the existing hyperpolarized (HP) 13C MRI analyses use univariate rate maps of pyruvate-to-lactate conversion (kPL), and radiomic-style multiparametric models extracting complex, higher-order features remain unexplored. Purpose: To establish a multivariate framework based on whole abdomen/pelvis HP 13 [...] Read more.
Background: Most of the existing hyperpolarized (HP) 13C MRI analyses use univariate rate maps of pyruvate-to-lactate conversion (kPL), and radiomic-style multiparametric models extracting complex, higher-order features remain unexplored. Purpose: To establish a multivariate framework based on whole abdomen/pelvis HP 13C-pyruvate MRI and evaluate the association between multiparametric features of metabolism (MFM) and clinical outcome measures in advanced and metastatic prostate cancer. Methods: Retrospective statistical analysis was performed on 16 participants with metastatic or local-regionally advanced prostate cancer prospectively enrolled in a tertiary center who underwent HP-pyruvate MRI of abdomen or pelvis between November 2020 and May 2023. Five patients were hormone-sensitive and eleven were castration-resistant. GMP-grade [1-13C]pyruvate was polarized using a 5T clinical-research DNP polarizer, and HP MRI used a set of flexible vest-transmit, array-receive coils, and echo-planar imaging sequences. Three basic metabolic maps (kPL, pyruvate summed-over-time, and mean pyruvate time) were created by semi-automatic segmentation, from which 316 MFMs were extracted using an open-source, radiomic-compliant software package. Univariate risk classifier was constructed using a biologically meaningful feature (kPL,median), and the multivariate classifier used a two-step feature selection process (ranking and clustering). Both were correlated with progression-free survival (PFS) and overall survival (OS) (median follow-up = 22.0 months) using Cox proportional hazards model. Results: In the univariate analysis, patients harboring tumors with lower-kPL,median had longer PFS (11.2 vs. 0.5 months, p < 0.01) and OS (NR vs. 18.4 months, p < 0.05) than their higher-kPL,median counterparts. Using a hypothesis-generating, age-adjusted multivariate risk classifier, the lower-risk subgroup also had longer PFS (NR vs. 2.4 months, p < 0.002) and OS (NR vs. 18.4 months, p < 0.05). By contrast, established laboratory markers, including PSA, lactate dehydrogenase, and alkaline phosphatase, were not significantly associated with PFS or OS (p > 0.05). Key limitations of this study include small sample size, retrospective study design, and referral bias. Conclusions: Risk classifiers derived from select multiparametric HP features were significantly associated with clinically meaningful outcome measures in this small, heterogeneous patient cohort, strongly supporting further investigation into their prognostic values. Full article
Show Figures

Figure 1

19 pages, 1827 KB  
Article
ISUP Grade Prediction of Prostate Nodules on T2WI Acquisitions Using Clinical Features, Textural Parameters and Machine Learning-Based Algorithms
by Teodora Telecan, Alexandra Chiorean, Roxana Sipos-Lascu, Cosmin Caraiani, Bianca Boca, Raluca Maria Hendea, Teodor Buliga, Iulia Andras, Nicolae Crisan and Monica Lupsor-Platon
Cancers 2025, 17(12), 2035; https://doi.org/10.3390/cancers17122035 - 18 Jun 2025
Cited by 3 | Viewed by 823
Abstract
Background: Prostate cancer (PCa) represents a matter at the forefront of healthcare, being divided into clinically significant (csPCa) and indolent PCa based on prognostic and treatment options. Although multi-parametric magnetic resonance imaging (mpMRI) has enabled significant advances, it cannot differentiate between the aforementioned [...] Read more.
Background: Prostate cancer (PCa) represents a matter at the forefront of healthcare, being divided into clinically significant (csPCa) and indolent PCa based on prognostic and treatment options. Although multi-parametric magnetic resonance imaging (mpMRI) has enabled significant advances, it cannot differentiate between the aforementioned categories; therefore, in order to render the initial diagnosis, invasive procedures such as transrectal prostate biopsy are still necessary. In response to these challenges, artificial intelligence (AI)-based algorithms combined with radiomics features offer the possibility of creating a textural pixel pattern-based surrogate, which has the potential of correlating the medical imagery with the pathological report in a one-to-one manner. Objective: The aim of the present study was to develop a machine learning model that can differentiate indolent from csPCa lesions, as well as individually classifying each nodule into corresponding ISUP grades prior to prostate biopsy, using textural features derived from mpMRI T2WI acquisitions. Materials and Methods: The study was conducted in 154 patients and 201 individual prostatic lesions. All cases were scanned using the same 1.5 Tesla mpMRI machine, employing a standard protocol. Each nodule was manually delineated using the 3D Slicer platform (version 5.2.2) and textural parameters were derived using the PyRadiomics database (version 3.1.0). We compared three machine learning classification models (Random Forest, Support Vector Machine, and Logistic Regression) in full, partial and no correlation settings, in order to differentiate between indolent and csPCa, as well as between ISUP 2 and ISUP 3 lesions. Results: The median age was 65 years (IQR: 61–69), the mean PSA value was 10.27 ng/mL, and 76.61% of the segmented lesions had a PI-RADS score of 4 or higher. Overall, the highest performance was registered for the Random Forest model in the partial correlation setting, differentiating between indolent and csPCa and between ISUP 2 versus ISUP 3 lesions, with accuracies of 88.13% and 82.5%, respectively. When the models were trained on combined clinical data and radiomic signatures, these accuracies increased to 91.11% and 91.39%, respectively. Conclusions: We developed a machine learning decision support tool that accurately predicts the ISUP grade prior to prostate biopsy, based on the textural features extracted from T2 MRI acquisitions. Full article
(This article belongs to the Section Cancer Causes, Screening and Diagnosis)
Show Figures

Figure 1

Back to TopTop