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
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
remove_circle_outline
remove_circle_outline

Search Results (700)

Search Parameters:
Keywords = risk stratification systems

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
31 pages, 419 KiB  
Review
Neoadjuvant Treatment for Locally Advanced Rectal Cancer: Current Status and Future Directions
by Masayoshi Iwamoto, Kazuki Ueda and Junichiro Kawamura
Cancers 2025, 17(15), 2540; https://doi.org/10.3390/cancers17152540 - 31 Jul 2025
Viewed by 190
Abstract
Locally advanced rectal cancer (LARC) remains a major clinical challenge due to its high risk of local recurrence and distant metastasis. Although total mesorectal excision (TME) has been established as the gold standard surgical approach, high recurrence rates associated with surgery alone have [...] Read more.
Locally advanced rectal cancer (LARC) remains a major clinical challenge due to its high risk of local recurrence and distant metastasis. Although total mesorectal excision (TME) has been established as the gold standard surgical approach, high recurrence rates associated with surgery alone have driven the development of multimodal preoperative strategies, such as radiotherapy and chemoradiotherapy. More recently, total neoadjuvant therapy (TNT)—which integrates systemic chemotherapy and radiotherapy prior to surgery—and non-operative management (NOM) for patients who achieve a clinical complete response (cCR) have further expanded treatment options. These advances aim not only to improve oncologic outcomes but also to enhance quality of life (QOL) by reducing long-term morbidity and preserving organ function. However, several unresolved issues persist, including the optimal sequencing of therapies, precise risk stratification, accurate evaluation of treatment response, and effective surveillance protocols for NOM. The advent of molecular biomarkers, next-generation sequencing, and artificial intelligence (AI) presents new opportunities for individualized treatment and more accurate prognostication. This narrative review provides a comprehensive overview of the current status of preoperative treatment for LARC, critically examines emerging strategies and their supporting evidence, and discusses future directions to optimize both oncological and patient-centered outcomes. By integrating clinical, molecular, and technological advances, the management of rectal cancer is moving toward truly personalized medicine. Full article
(This article belongs to the Special Issue Multidisciplinary Management of Rectal Cancer)
17 pages, 812 KiB  
Article
Association Between ABO Blood Groups and SARS-CoV-2 RNAemia, Spike Protein Mutations, and Thrombotic Events in COVID-19 Patients
by Esra’a Abudouleh, Tarek Owaidah, Fatimah Alhamlan, Arwa A. Al-Qahtani, Dalia Al Sarar, Abdulrahman Alkathiri, Shouq Alghannam, Arwa Bagasi, Manal M. Alkhulaifi and Ahmed A. Al-Qahtani
Pathogens 2025, 14(8), 758; https://doi.org/10.3390/pathogens14080758 (registering DOI) - 31 Jul 2025
Viewed by 103
Abstract
Background: COVID-19 is associated with coagulopathy and increased mortality. The ABO blood group system has been implicated in modulating susceptibility to SARS-CoV-2 infection and disease severity, but its relationship with viral RNAemia, spike gene mutations, and thrombosis remains underexplored. Methods: We analyzed 446 [...] Read more.
Background: COVID-19 is associated with coagulopathy and increased mortality. The ABO blood group system has been implicated in modulating susceptibility to SARS-CoV-2 infection and disease severity, but its relationship with viral RNAemia, spike gene mutations, and thrombosis remains underexplored. Methods: We analyzed 446 hospitalized COVID-19 patients between 2021 and 2022. SARS-CoV-2 RNAemia was assessed via RT-qPCR on whole blood, and spike gene mutations were identified through whole-genome sequencing in RNAemia-positive samples. ABO blood groups were determined by agglutination testing, and thrombotic events were evaluated using coagulation markers. Statistical analyses included chi-square tests and Kruskal–Wallis tests, with significance set at p < 0.05. Results: RNAemia was detected in 26.9% of patients, with no significant association with ABO blood group (p = 0.175). Omicron was the predominant variant, especially in blood group A (62.5%). The N501Y mutation was the most prevalent in group O (53.2%), and K417N was most prevalent in group B (36.9%), though neither reached statistical significance. Thrombotic events were significantly more common in blood group A (OR = 2.08, 95% CI = 1.3–3.4, p = 0.002), particularly among RNAemia-positive patients. Conclusions: ABO blood group phenotypes, particularly group A, may influence thrombotic risk in the context of SARS-CoV-2 RNAemia. While no direct association was found between blood group and RNAemia or spike mutations, the observed trends suggest potential host–pathogen interactions. Integrating ABO typing and RNAemia screening may enhance risk stratification and guide targeted thromboprophylaxis in COVID-19 patients. Full article
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 81
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

24 pages, 728 KiB  
Review
Pathophysiological Links Between Inflammatory Bowel Disease and Cardiovascular Disease: The Role of Dysbiosis and Emerging Biomarkers
by Roko Šantić, Nikola Pavlović, Marko Kumrić, Marino Vilović and Joško Božić
Biomedicines 2025, 13(8), 1864; https://doi.org/10.3390/biomedicines13081864 - 31 Jul 2025
Viewed by 84
Abstract
This review introduces a novel integrative framework linking gut dysbiosis, systemic inflammation, and cardiovascular risk in patients with inflammatory bowel disease (IBD). We highlight emerging biomarkers, including short-chain fatty acids (SCFAs), calprotectin, and zonulin, that reflect alterations in the gut microbiome and increased [...] Read more.
This review introduces a novel integrative framework linking gut dysbiosis, systemic inflammation, and cardiovascular risk in patients with inflammatory bowel disease (IBD). We highlight emerging biomarkers, including short-chain fatty acids (SCFAs), calprotectin, and zonulin, that reflect alterations in the gut microbiome and increased intestinal permeability, which contribute to cardiovascular pathology. Cardiovascular diseases (CVDs) remain the leading cause of morbidity and mortality worldwide, and recent evidence identifies IBD, encompassing ulcerative colitis (UC) and Crohn’s disease (CD), as a significant non-traditional risk factor for CVD. This review synthesizes current knowledge on how dysbiosis-driven inflammation in IBD patients exacerbates endothelial dysfunction, hypercoagulability, and atherosclerosis, even in the absence of traditional risk factors. Additionally, we discuss how commonly used IBD therapies may modulate cardiovascular risk. Understanding these multifactorial mechanisms and validating reliable biomarkers are essential for improving cardiovascular risk stratification and guiding targeted prevention strategies in this vulnerable population. Full article
15 pages, 2220 KiB  
Article
Radiologic Assessment of Periprostatic Fat as an Indicator of Prostate Cancer Risk on Multiparametric MRI
by Roxana Iacob, Emil Radu Iacob, Emil Robert Stoicescu, Diana Manolescu, Laura Andreea Ghenciu, Radu Căprariu, Amalia Constantinescu, Iulia Ciobanu, Răzvan Bardan and Alin Cumpănaș
Bioengineering 2025, 12(8), 831; https://doi.org/10.3390/bioengineering12080831 (registering DOI) - 31 Jul 2025
Viewed by 149
Abstract
Prostate cancer remains one of the most prevalent malignancies among men, and emerging evidence proposed a potential role for periprostatic adipose tissue (PPAT) in tumor progression. However, its relationship with imaging-based risk stratification systems such as PI-RADS remains uncertain. This retrospective observational study [...] Read more.
Prostate cancer remains one of the most prevalent malignancies among men, and emerging evidence proposed a potential role for periprostatic adipose tissue (PPAT) in tumor progression. However, its relationship with imaging-based risk stratification systems such as PI-RADS remains uncertain. This retrospective observational study aimed to evaluate whether periprostatic and subcutaneous fat thickness are associated with PI-RADS scores or PSA levels in biopsy-naïve patients. We retrospectively reviewed 104 prostate MRI scans performed between January 2020 and January 2024. Fat thickness was measured on axial T2-weighted images, and statistical analyses were conducted using Spearman’s correlation and multiple linear regression. In addition to linear measurements, we also assessed periprostatic fat volume and posterior fat thickness derived from imaging data. No significant correlations were observed between fat thickness (either periprostatic or subcutaneous) and PI-RADS score or PSA values. Similarly, periprostatic fat volume showed only a weak, non-significant correlation with PI-RADS, while posterior fat thickness demonstrated a weak but statistically significant positive association. Additionally, subgroup comparisons between low-risk (PI-RADS < 4) and high-risk (PI-RADS ≥ 4) patients showed no meaningful differences in fat measurements. These findings suggest that simple linear fat thickness measurements may not enhance imaging-based risk assessment in prostate cancer, though regional and volumetric assessments could offer modest added value. Full article
(This article belongs to the Special Issue Label-Free Cancer Detection)
Show Figures

Figure 1

16 pages, 929 KiB  
Article
Galectin-3 Reflects Systemic Atherosclerosis in Patients with Coronary Artery Disease
by Horea-Laurentiu Onea, Calin Homorodean, Florin-Leontin Lazar, Mihai Octavian Negrea, Teodora Calin, Ioan Cornel Bitea, Minodora Teodoru, Vlad Ionut Nechita, Ariela Ligia Olteanu and Dan-Mircea Olinic
Medicina 2025, 61(8), 1388; https://doi.org/10.3390/medicina61081388 - 30 Jul 2025
Viewed by 165
Abstract
Background and Objectives: Galectin-3 (Gal-3), a pro-inflammatory cytokine, has been implicated in atherosclerosis and adverse cardiovascular outcomes. While its role in coronary artery disease (CAD) is increasingly recognized, its association with systemic atherosclerosis remains underexplored. Objective: To investigate serum Gal-3 levels in [...] Read more.
Background and Objectives: Galectin-3 (Gal-3), a pro-inflammatory cytokine, has been implicated in atherosclerosis and adverse cardiovascular outcomes. While its role in coronary artery disease (CAD) is increasingly recognized, its association with systemic atherosclerosis remains underexplored. Objective: To investigate serum Gal-3 levels in patients with CAD and evaluate correlations between CAD severity and extra-coronary atherosclerotic involvement (carotid, femoral, and radial territories). Materials and Methods: We prospectively enrolled 56 patients with CAD undergoing coronary angiography (42.8% with acute-ACS; 57.2% with chronic coronary syndromes-CCS). Gal-3 levels were measured within 24 h of admission. Atherosclerosis severity was assessed angiographically and through vascular ultrasound of the carotid, femoral, and radial arteries. Patients were stratified by median Gal-3 levels, and clinical follow-up was performed at 1 and 3 months. Results: Gal-3 levels were significantly higher in CAD vs. controls (20.7 vs. 10.1 ng/mL; p < 0.00001) and in ACS vs. CCS (22.18. vs. 17.93 ng/mL; p = 0.019). Gal-3 correlated positively with culprit lesion diameter stenosis (DS) (R = 0.30; p = 0.023) and maximum severity of additional treated lesions (R = 0.62; p = 0.006). Gal-3 also correlated positively with carotid plaque thickness (R = 0.32; p = 0.016), while patients with Gal-3 levels above the median showed increased median values for femoral plaque thickness (32.4 vs. 26.45 mm, p = 0.046). No correlation was found with radial artery calcification. Gal-3 showed moderate discrimination for ACS (AUC = 0.685; cut-off 20.18 ng/mL). On multivariate analysis age, DS, and ACS presentation were independent predictors of Gal-3 above 19.07 ng/mL. Conclusions: Gal-3 levels are elevated in ACS and correlate with atherosclerotic burden, particularly in coronary, carotid, and femoral territories. These findings support Gal-3 as a potential marker of lesion severity and systemic vascular involvement, highlighting its possible role in risk stratification and the monitoring of atherosclerotic disease progression. This study provides integrated insights into the impact of Gal-3 across multiple vascular beds by assessing them concurrently within the same patient cohort. Full article
(This article belongs to the Section Cardiology)
Show Figures

Graphical abstract

12 pages, 456 KiB  
Article
From Variability to Standardization: The Impact of Breast Density on Background Parenchymal Enhancement in Contrast-Enhanced Mammography and the Need for a Structured Reporting System
by Graziella Di Grezia, Antonio Nazzaro, Luigi Schiavone, Cisternino Elisa, Alessandro Galiano, Gatta Gianluca, Cuccurullo Vincenzo and Mariano Scaglione
Cancers 2025, 17(15), 2523; https://doi.org/10.3390/cancers17152523 - 30 Jul 2025
Viewed by 318
Abstract
Introduction: Breast density is a well-recognized factor in breast cancer risk assessment, with higher density linked to increased malignancy risk and reduced sensitivity of conventional mammography. Background parenchymal enhancement (BPE), observed in contrast-enhanced imaging, reflects physiological contrast uptake in non-pathologic breast tissue. [...] Read more.
Introduction: Breast density is a well-recognized factor in breast cancer risk assessment, with higher density linked to increased malignancy risk and reduced sensitivity of conventional mammography. Background parenchymal enhancement (BPE), observed in contrast-enhanced imaging, reflects physiological contrast uptake in non-pathologic breast tissue. While extensively characterized in breast MRI, the role of BPE in contrast-enhanced mammography (CEM) remains uncertain due to inconsistent findings regarding its correlation with breast density and cancer risk. Unlike breast density—standardized through the ACR BI-RADS lexicon—BPE lacks a uniform classification system in CEM, leading to variability in clinical interpretation and research outcomes. To address this gap, we introduce the BPE-CEM Standard Scale (BCSS), a structured four-tiered classification system specifically tailored to the two-dimensional characteristics of CEM, aiming to improve consistency and diagnostic alignment in BPE evaluation. Materials and Methods: In this retrospective single-center study, 213 patients who underwent mammography (MG), ultrasound (US), and contrast-enhanced mammography (CEM) between May 2022 and June 2023 at the “A. Perrino” Hospital in Brindisi were included. Breast density was classified according to ACR BI-RADS (categories A–D). BPE was categorized into four levels: Minimal (< 10% enhancement), Light (10–25%), Moderate (25–50%), and Marked (> 50%). Three radiologists independently assessed BPE in a subset of 50 randomly selected cases to evaluate inter-observer agreement using Cohen’s kappa. Correlations between BPE, breast density, and age were examined through regression analysis. Results: BPE was Minimal in 57% of patients, Light in 31%, Moderate in 10%, and Marked in 2%. A significant positive association was found between higher breast density (BI-RADS C–D) and increased BPE (p < 0.05), whereas lower-density breasts (A–B) were predominantly associated with minimal or light BPE. Regression analysis confirmed a modest but statistically significant association between breast density and BPE (R2 = 0.144), while age showed no significant effect. Inter-observer agreement for BPE categorization using the BCSS was excellent (κ = 0.85; 95% CI: 0.78–0.92), supporting its reproducibility. Conclusions: Our findings indicate that breast density is a key determinant of BPE in CEM. The proposed BCSS offers a reproducible, four-level framework for standardized BPE assessment tailored to the imaging characteristics of CEM. By reducing variability in interpretation, the BCSS has the potential to improve diagnostic consistency and facilitate integration of BPE into personalized breast cancer risk models. Further prospective multicenter studies are needed to validate this classification and assess its clinical impact. Full article
Show Figures

Figure 1

15 pages, 1343 KiB  
Article
Prognostic Value of Metabolic Tumor Volume and Heterogeneity Index in Diffuse Large B-Cell Lymphoma
by Ali Alper Solmaz, Ilhan Birsenogul, Aygul Polat Kelle, Pinar Peker, Burcu Arslan Benli, Serdar Ata, Mahmut Bakir Koyuncu, Mustafa Gurbuz, Ali Ogul, Berna Bozkurt Duman and Timucin Cil
Medicina 2025, 61(8), 1370; https://doi.org/10.3390/medicina61081370 - 29 Jul 2025
Viewed by 468
Abstract
Background and Objectives: Metabolic tumor volume (MTV) and inflammation-based indices have recently gained attention as potential prognostic markers of diffuse large B-cell lymphoma (DLBCL). We aimed to evaluate the prognostic significance of metabolic and systemic inflammatory parameters in predicting treatment response, relapse, [...] Read more.
Background and Objectives: Metabolic tumor volume (MTV) and inflammation-based indices have recently gained attention as potential prognostic markers of diffuse large B-cell lymphoma (DLBCL). We aimed to evaluate the prognostic significance of metabolic and systemic inflammatory parameters in predicting treatment response, relapse, and overall survival (OS) in patients with DLBCL. Materials and Methods: This retrospective cohort study included 70 patients with DLBCL. Clinical characteristics, laboratory values, and metabolic parameters, including maximum standardized uptake value (SUVmaxliver and SUVmax), heterogeneity indices HI1 and HI2, and MTV were analyzed. Survival outcomes were assessed using Kaplan–Meier and log-rank tests. Receiver operating characteristic analyses helped evaluate the diagnostic performance of the selected biomarkers in predicting relapse and mortality. Univariate and multivariate logistic regression analyses were conducted to identify the independent predictors. Results: The mean OS and mean relapse-free survival (RFS) were 71.6 ± 7.4 and 38.7 ± 2.9 months, respectively. SUVmaxliver ≤ 22 and HI2 > 62.3 were associated with a significantly shorter OS. High lactate dehydrogenase (LDH) levels and HI2 > 87.9 were significantly associated with a reduced RFS. LDH, SUVmaxliver, and HI2 had a significant predictive value for relapse. SUVmaxliver and HI2 levels were also predictive of mortality; SUVmaxliver ≤ 22 and HI2 > 62.3 independently predicted mortality, while HI2 > 87.9 independently predicted relapse. MTV was not significantly associated with survival. Conclusions: Metabolic tumor burden and inflammation-based markers, particularly SUVmaxliver and HI2, are significant prognostic indicators of DLBCL and may enhance risk stratification and aid in identifying patients with an increased risk of relapse or mortality, potentially guiding personalized therapy. Full article
(This article belongs to the Section Oncology)
Show Figures

Figure 1

14 pages, 1712 KiB  
Article
Machine Learning-Based Predictive Model for Risk Stratification of Multiple Myeloma from Monoclonal Gammopathy of Undetermined Significance
by Amparo Santamaría, Marcos Alfaro, Cristina Antón, Beatriz Sánchez-Quiñones, Nataly Ibarra, Arturo Gil, Oscar Reinoso and Luis Payá
Electronics 2025, 14(15), 3014; https://doi.org/10.3390/electronics14153014 - 29 Jul 2025
Viewed by 239
Abstract
Monoclonal Gammopathy of Undetermined Significance (MGUS) is a precursor to hematologic malignancies such as Multiple Myeloma (MM) and Waldenström Macroglobulinemia (WM). Accurate risk stratification of MGUS patients remains a clinical and computational challenge, with existing models often misclassifying both high-risk and low-risk individuals, [...] Read more.
Monoclonal Gammopathy of Undetermined Significance (MGUS) is a precursor to hematologic malignancies such as Multiple Myeloma (MM) and Waldenström Macroglobulinemia (WM). Accurate risk stratification of MGUS patients remains a clinical and computational challenge, with existing models often misclassifying both high-risk and low-risk individuals, leading to inefficient healthcare resource allocation. This study presents a machine learning (ML)-based approach for early prediction of MM/WM progression, using routinely collected hematological data, which are selected based on clinical relevance. A retrospective cohort of 292 MGUS patients, including 7 who progressed to malignancy, was analyzed. For each patient, a feature descriptor was constructed incorporating the latest biomarker values, their temporal trends over the previous year, age, and immunoglobulin subtype. To address the inherent class imbalance, data augmentation techniques were applied. Multiple ML classifiers were evaluated, with the Support Vector Machine (SVM) achieving the highest performance (94.3% accuracy and F1-score). The model demonstrates that a compact set of clinically relevant features can yield robust predictive performance. These findings highlight the potential of ML-driven decision-support systems in electronic health applications, offering a scalable solution for improving MGUS risk stratification, optimizing clinical workflows, and enabling earlier interventions. Full article
Show Figures

Graphical abstract

19 pages, 766 KiB  
Systematic Review
Molecular Mechanisms Underlying Inflammation in Early-Onset Neonatal Sepsis: A Systematic Review of Human Studies
by Anca Vulcănescu, Mirela-Anișoara Siminel, Anda-Lorena Dijmărescu, Maria-Magdalena Manolea, Sidonia-Maria Săndulescu, Virginia Maria Rădulescu, Valeriu Gheorman and Sorin-Nicolae Dinescu
J. Clin. Med. 2025, 14(15), 5315; https://doi.org/10.3390/jcm14155315 - 28 Jul 2025
Viewed by 283
Abstract
Background/Objective: Early-onset neonatal sepsis (EOS), defined as infection occurring within the first 72 h after birth, remains a major contributor to neonatal morbidity and mortality worldwide. Although advances in perinatal care have improved overall outcomes, the diagnosis of EOS continues to be [...] Read more.
Background/Objective: Early-onset neonatal sepsis (EOS), defined as infection occurring within the first 72 h after birth, remains a major contributor to neonatal morbidity and mortality worldwide. Although advances in perinatal care have improved overall outcomes, the diagnosis of EOS continues to be challenging. Clinical presentations are often nonspecific, laboratory confirmation is often delayed, and immune responses vary considerably among neonates. Expanding our understanding of the molecular mechanisms underlying EOS is essential in enhancing early detection, refining risk stratification, and guiding therapeutic strategies. This systematic review aims to synthesize the available information on the molecular pathways involved in EOS, focusing on pathogen-induced inflammation, systemic immune responses, sterile inflammatory processes, interactions between infectious and non-infectious pathways, as well as emerging molecular diagnostic approaches. Methods: A comprehensive review of original research articles and reviews published between January 2015 and January 2025 was conducted; studies were included based on their focus on human neonates and their analysis of molecular or immunological mechanisms relevant to EOS pathogenesis, immune dysregulation, or novel diagnostic strategies. Results: Pathogen-driven inflammation typically involves the activation of Toll-like receptors (TLRs), the recruitment of neutrophils, and the release of pro-inflammatory cytokines such as IL-6, IL-1β, and TNF-α, particularly in response to vertical transmission of organisms like Escherichia coli and Streptococcus agalactiae. Systemic inflammatory responses are marked by cytokine dysregulation, contributing to multi-organ dysfunction. Sterile inflammation, often initiated by hypoxia–reperfusion injury or intrauterine stress, amplifies susceptibility to sepsis. Interactions between immune, metabolic, and endothelial pathways further exacerbate tissue injury. Recent advances, including transcriptomic profiling, microRNA-based biomarkers, and immune checkpoint studies, offer promising strategies for earlier diagnosis and individualized therapeutic options. Conclusions: EOS arises from a complex interplay of infectious and sterile inflammatory mechanisms. A deeper molecular understanding holds promise for advancing correct diagnostics and targeted therapies, aiming to improve neonatal outcomes. Full article
(This article belongs to the Section Clinical Pediatrics)
Show Figures

Figure 1

23 pages, 1324 KiB  
Review
Advances and Challenges in the Management of Myelodysplastic Syndromes
by Jessica M. Stempel, Tariq Kewan and Amer M. Zeidan
Cancers 2025, 17(15), 2469; https://doi.org/10.3390/cancers17152469 - 25 Jul 2025
Viewed by 890
Abstract
Myelodysplastic syndromes/neoplasms (MDS) represent a biologically and clinically diverse group of myeloid malignancies marked by cytopenias, morphological dysplasia, and an inherent risk of progression to acute myeloid leukemia. Over the past two decades, the field has made significant advances in characterizing the molecular [...] Read more.
Myelodysplastic syndromes/neoplasms (MDS) represent a biologically and clinically diverse group of myeloid malignancies marked by cytopenias, morphological dysplasia, and an inherent risk of progression to acute myeloid leukemia. Over the past two decades, the field has made significant advances in characterizing the molecular landscape of MDS, leading to refined classification systems to reflect the underlying genetic and biological diversity. In 2025, the treatment of MDS is increasingly individualized, guided by integrated clinical, cytogenetic, and molecular risk stratification tools. For lower-risk MDS, the treatment paradigm has evolved beyond erythropoiesis-stimulating agents (ESAs) with the introduction of novel effective agents such as luspatercept and imetelstat, as well as shortened schedules of hypomethylating agents (HMAs). For higher-risk disease, monotherapy with HMAs continue to be the standard of care as combination therapies of HMAs with novel agents have, to date, failed to redefine treatment paradigms. The recognition of precursor states like clonal hematopoiesis of indeterminate potential (CHIP) and the increasing use of molecular monitoring will hopefully enable earlier intervention/prevention strategies. This review provides a comprehensive overview of the current treatment approach for MDS, highlighting new classifications, prognostic tools, evolving therapeutic options, and ongoing challenges. We discuss evidence-based recommendations, treatment sequencing, and emerging clinical trials, with a focus on translating biological insights into improved outcomes for patients with MDS. Full article
(This article belongs to the Special Issue New Insights of Hematology in Cancer)
Show Figures

Figure 1

13 pages, 1452 KiB  
Article
Prognostic Utility of Combining VI-RADS Scores and CYFRA 21-1 Levels in Bladder Cancer: A Retrospective Single-Center Study
by Shunsuke Ikuma, Jun Akatsuka, Godai Kaneko, Hayato Takeda, Yuki Endo, Go Kimura and Yukihiro Kondo
Curr. Oncol. 2025, 32(8), 415; https://doi.org/10.3390/curroncol32080415 - 24 Jul 2025
Viewed by 232
Abstract
The Vesical Imaging Reporting and Data System (VI-RADS) is used to detect muscle-invasive bladder cancer, with emerging prognostic implications. Integrating imaging parameters with molecular biomarkers may improve risk stratification in bladder cancer. This study evaluated whether combining VI-RADS scores with serum cytokeratin fragment [...] Read more.
The Vesical Imaging Reporting and Data System (VI-RADS) is used to detect muscle-invasive bladder cancer, with emerging prognostic implications. Integrating imaging parameters with molecular biomarkers may improve risk stratification in bladder cancer. This study evaluated whether combining VI-RADS scores with serum cytokeratin fragment 19 (CYFRA 21-1) levels—a clinically relevant biomarker for bladder cancer—could improve overall survival (OS) prediction. We retrospectively analyzed 134 patients who underwent transurethral resection of bladder tumors, magnetic resonance imaging, and postoperative serum CYFRA 21-1 measurements. In total, 15 cancer-specific deaths were observed during follow-up. Receiver operating characteristic curve analysis identified optimal prognostic cut-off values: VI-RADS score ≥ 4 and CYFRA 21-1 level ≥ 1.8 ng/mL. The 1-, 2-, and 3-year OS in patients with both high VI-RADS scores and CYFRA 21-1 levels were 42.9%, 16.7%, and 8.3%, respectively, significantly lower than those in other groups (p < 0.001, 0.002, and 0.003, respectively). Multivariate Cox proportional hazards analysis demonstrated that such patients had the poorest OS (hazard ratio: 7.51; p = 0.002). This suggests that combining VI-RADS and CYFRA 21-1 improves prognostic accuracy in bladder cancer, demonstrating potential clinical utility by informing individualized treatment strategies; however, limitations include the retrospective study design and absence of external validation. Full article
(This article belongs to the Section Genitourinary Oncology)
Show Figures

Graphical abstract

13 pages, 2372 KiB  
Article
PTEN and ERG Biomarkers as Predictors of Biochemical Recurrence Risk in Patients Undergoing Radical Prostatectomy
by Mihnea Bogdan Borz, Bogdan Fetica, Maximilian Cosma Gliga, Tamas-Csaba Sipos, Bogdan Adrian Buhas and Vlad Horia Schitcu
Diseases 2025, 13(8), 235; https://doi.org/10.3390/diseases13080235 - 24 Jul 2025
Viewed by 260
Abstract
Background/Objectives: Prostate cancer (PCa) remains a major global health issue, associated with significant mortality and morbidity. Despite advances in diagnosis and treatment, predicting biochemical recurrence (BCR) after radical prostatectomy remains challenging, highlighting the need for reliable biomarkers to guide prognosis and therapy. [...] Read more.
Background/Objectives: Prostate cancer (PCa) remains a major global health issue, associated with significant mortality and morbidity. Despite advances in diagnosis and treatment, predicting biochemical recurrence (BCR) after radical prostatectomy remains challenging, highlighting the need for reliable biomarkers to guide prognosis and therapy. The study aimed to evaluate the prognostic significance of the PTEN and ERG biomarkers in predicting BCR and tumor progression in PCa patients who underwent radical prostatectomy. Methods: This study consisted of a cohort of 91 patients with localized PCa who underwent radical prostatectomy between 2016 and 2022. From this cohort, 77 patients were selected for final analysis. Tissue microarrays (TMAs) were constructed from paraffin blocks, and immunohistochemical (IHC) staining for PTEN and ERG was performed using specific antibodies on the Ventana BenchMark ULTRA system (Roche Diagnostics, Indianapolis, IN, USA). Stained sections were evaluated and correlated with clinical and pathological data. Results: PTEN expression showed a significant negative correlation with BCR (r = −0.301, p = 0.014), indicating that reduced PTEN expression is associated with increased recurrence risk. PTEN was not significantly linked to PSA levels, tumor stage, or lymph node involvement. ERG expression correlated positively with advanced pathological tumor stage (r = 0.315, p = 0.005) but was not associated with BCR or other clinical parameters. Conclusions: PTEN appears to be a valuable prognostic marker for recurrence in PCa, while ERG may indicate tumor progression. These findings support the potential integration of PTEN and ERG into clinical practice to enhance risk stratification and personalized treatment, warranting further validation in larger patient cohorts. Full article
(This article belongs to the Section Oncology)
Show Figures

Figure 1

19 pages, 967 KiB  
Review
Hematologic and Immunologic Overlap Between COVID-19 and Idiopathic Pulmonary Fibrosis
by Gabriela Mara, Gheorghe Nini, Stefan Marian Frenț and Coralia Cotoraci
J. Clin. Med. 2025, 14(15), 5229; https://doi.org/10.3390/jcm14155229 - 24 Jul 2025
Viewed by 341
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive fibrosing lung disease characterized by chronic inflammation, vascular remodeling, and immune dysregulation. COVID-19, caused by SARS-CoV-2, shares several systemic immunohematologic disturbances with IPF, including cytokine storms, endothelial injury, and prothrombotic states. Unlike general comparisons of viral [...] Read more.
Idiopathic pulmonary fibrosis (IPF) is a progressive fibrosing lung disease characterized by chronic inflammation, vascular remodeling, and immune dysregulation. COVID-19, caused by SARS-CoV-2, shares several systemic immunohematologic disturbances with IPF, including cytokine storms, endothelial injury, and prothrombotic states. Unlike general comparisons of viral infections and chronic lung disease, this review offers a focused analysis of the shared hematologic and immunologic mechanisms between COVID-19 and IPF. Our aim is to better understand how SARS-CoV-2 infection may worsen disease progression in IPF and identify converging pathophysiological pathways that may inform clinical management. We conducted a narrative synthesis of the peer-reviewed literature from PubMed, Scopus, and Web of Science, focusing on clinical, experimental, and pathological studies addressing immune and coagulation abnormalities in both COVID-19 and IPF. Both diseases exhibit significant overlap in inflammatory and fibrotic signaling, particularly via the TGF-β, IL-6, and TNF-α pathways. COVID-19 amplifies coagulation disturbances and endothelial dysfunction already present in IPF, promoting microvascular thrombosis and acute exacerbations. Myeloid cell overactivation, impaired lymphocyte responses, and fibroblast proliferation are central to this shared pathophysiology. These synergistic mechanisms may accelerate fibrosis and increase mortality risk in IPF patients infected with SARS-CoV-2. This review proposes an integrative framework for understanding the hematologic and immunologic convergence of COVID-19 and IPF. Such insights are essential for refining therapeutic targets, improving prognostic stratification, and guiding early interventions in this high-risk population. Full article
(This article belongs to the Special Issue Chronic Lung Conditions: Integrative Approaches to Long-Term Care)
Show Figures

Figure 1

47 pages, 4589 KiB  
Review
Understanding Sex Differences in Autoimmune Diseases: Immunologic Mechanisms
by Yu Rin Kim, YunJae Jung, Insug Kang and Eui-Ju Yeo
Int. J. Mol. Sci. 2025, 26(15), 7101; https://doi.org/10.3390/ijms26157101 - 23 Jul 2025
Viewed by 344
Abstract
Autoimmune diseases such as systemic lupus erythematosus and Sjögren’s syndrome show pronounced sex disparities in prevalence, severity, and clinical outcomes, with females disproportionately affected. Emerging evidence highlights sex-based differences in immune and inflammatory responses as key contributors to this bias. Genetic factors—including sex [...] Read more.
Autoimmune diseases such as systemic lupus erythematosus and Sjögren’s syndrome show pronounced sex disparities in prevalence, severity, and clinical outcomes, with females disproportionately affected. Emerging evidence highlights sex-based differences in immune and inflammatory responses as key contributors to this bias. Genetic factors—including sex chromosomes, skewed X chromosome inactivation, and sex-biased microRNAs—as well as sex hormones and pregnancy modulate gene expression and immune cell function in a sex-specific manner. Additionally, sex hormone-dependent epigenetic modifications influence the transcription of critical immune regulators. These genetic and hormonal factors collectively shape the activation, differentiation, and effector functions of diverse immune cell types. Environmental factors—including infections, gut microbiota, environmental chemicals and pollutants, and lifestyle behaviors such as diet, smoking, UV exposure, alcohol and caffeine intake, physical activity, and circadian rhythms—further modulate immune function and autoimmune disease pathogenesis in a sex-dependent manner. Together, these mechanisms contribute to the heightened risk and distinct clinical features of autoimmunity in females. A deeper understanding of sex-biased immune regulation will facilitate the identification of novel biomarkers, enable patient stratification, and inform the development of sex-specific diagnostic and therapeutic strategies for autoimmune diseases. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

Back to TopTop