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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (313)

Search Parameters:
Keywords = novel blood-based biomarker

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
37 pages, 1469 KiB  
Review
Oncolytic Therapies for Glioblastoma: Advances, Challenges, and Future Perspectives
by Omar Alomari, Habiba Eyvazova, Beyzanur Güney, Rana Al Juhmani, Hatice Odabasi, Lubna Al-Rawabdeh, Muhammed Edib Mokresh, Ufuk Erginoglu, Abdullah Keles and Mustafa K. Baskaya
Cancers 2025, 17(15), 2550; https://doi.org/10.3390/cancers17152550 - 1 Aug 2025
Viewed by 741
Abstract
Glioblastoma (GBM) remains one of the most aggressive and treatment-resistant brain tumors, necessitating novel therapeutic approaches. Oncolytic treatments, particularly oncolytic viruses (OVs), have emerged as promising candidates by selectively infecting and lysing tumor cells while stimulating anti-tumor immunity. Various virus-based therapies are under [...] Read more.
Glioblastoma (GBM) remains one of the most aggressive and treatment-resistant brain tumors, necessitating novel therapeutic approaches. Oncolytic treatments, particularly oncolytic viruses (OVs), have emerged as promising candidates by selectively infecting and lysing tumor cells while stimulating anti-tumor immunity. Various virus-based therapies are under investigation, including genetically engineered herpes simplex virus (HSV), adenovirus, poliovirus, reovirus, vaccinia virus, measles virus, and Newcastle disease virus, each exploiting unique tumor-selective mechanisms. While some, such as HSV-based therapies including G207 and DelytactTM, have demonstrated clinical progress, significant challenges persist, including immune evasion, heterogeneity in patient response, and delivery barriers due to the blood–brain barrier. Moreover, combination strategies integrating OVs with immune checkpoint inhibitors, chemotherapy, and radiation are promising but require further clinical validation. Non-viral oncolytic approaches, such as tumor-targeting bacteria and synthetic peptides, remain underexplored. This review highlights current advancements while addressing critical gaps in the literature, including the need for optimized delivery methods, better biomarker-based patient stratification, and a deeper understanding of GBM’s immunosuppressive microenvironment. Future research should focus on enhancing OV specificity, engineering viruses to deliver therapeutic genes, and integrating OVs with precision medicine strategies. By identifying these gaps, this review provides a framework for advancing oncolytic therapies in GBM treatment. Full article
(This article belongs to the Section Cancer Immunology and Immunotherapy)
Show Figures

Figure 1

14 pages, 882 KiB  
Article
Advancing Neonatal Screening for Pyridoxine-Dependent Epilepsy-ALDH7A1 Through Combined Analysis of 2-OPP, 6-Oxo-Pipecolate and Pipecolate in a Butylated FIA-MS/MS Workflow
by Mylène Donge, Sandrine Marie, Amandine Pochet, Lionel Marcelis, Geraldine Luis, François Boemer, Clément Prouteau, Samir Mesli, Matthias Cuykx, Thao Nguyen-Khoa, David Guénet, Aurélie Empain, Magalie Barth, Benjamin Dauriat, Cécile Laroche-Raynaud, Corinne De Laet, Patrick Verloo, An I. Jonckheere, Manuel Schiff, Marie-Cécile Nassogne and Joseph P. Dewulfadd Show full author list remove Hide full author list
Int. J. Neonatal Screen. 2025, 11(3), 59; https://doi.org/10.3390/ijns11030059 - 30 Jul 2025
Viewed by 306
Abstract
Pyridoxine-dependent epilepsy (PDE) represents a group of rare developmental and epileptic encephalopathies. The most common PDE is caused by biallelic pathogenic variants in ALDH7A1 (PDE-ALDH7A1; OMIM #266100), which encodes α-aminoadipate semialdehyde (α-AASA) dehydrogenase, a key enzyme in lysine catabolism. Affected individuals present with [...] Read more.
Pyridoxine-dependent epilepsy (PDE) represents a group of rare developmental and epileptic encephalopathies. The most common PDE is caused by biallelic pathogenic variants in ALDH7A1 (PDE-ALDH7A1; OMIM #266100), which encodes α-aminoadipate semialdehyde (α-AASA) dehydrogenase, a key enzyme in lysine catabolism. Affected individuals present with seizures unresponsive to conventional anticonvulsant medications but responsive to high-dose of pyridoxine (vitamin B6). Adjunctive lysine restriction and arginine supplementation have also shown potential in improving neurodevelopmental outcomes. Given the significant benefit of early intervention, PDE-ALDH7A1 is a strong candidate for newborn screening (NBS). However, traditional biomarkers are biochemically unstable at room temperature (α-AASA and piperideine-6-carboxylate) or lack sufficient specificity (pipecolate), limiting their utility for biomarker-based NBS. The recent identification of two novel and stable biomarkers, 2S,6S-/2S,6R-oxopropylpiperidine-2-carboxylate (2-OPP) and 6-oxo-pipecolate (oxo-PIP), offers renewed potential for biochemical NBS. We evaluated the feasibility of incorporating 2-OPP, oxo-PIP, and pipecolate into routine butylated FIA-MS/MS workflows used for biochemical NBS. A total of 9402 dried blood spots (DBS), including nine confirmed PDE-ALDH7A1 patients and 9393 anonymized controls were analyzed using a single multiplex assay. 2-OPP emerged as the most sensitive biomarker, identifying all PDE-ALDH7A1 patients with 100% sensitivity and a positive predictive value (PPV) of 18.4% using a threshold above the 99.5th percentile. Combining elevated 2-OPP (above the 99.5th percentile) with either pipecolate or oxo-PIP (above the 85.0th percentile) as secondary marker detected within the same multiplex FIA-MS/MS assay further improved the PPVs to 60% and 45%, respectively, while maintaining compatibility with butanol-derivatized method. Notably, increasing the 2-OPP threshold above the 99.89th percentile, in combination with either pipecolate or oxo-PIP above the 85.0th percentile resulted in both 100% sensitivity and 100% PPV. This study supports the strong potential of 2-OPP-based neonatal screening for PDE-ALDH7A1 within existing NBS infrastructures. The ability to multiplex 2-OPP, pipecolate and oxo-PIP within a single assay offers a robust, practical, high-throughput and cost-effective approach. These results support the inclusion of PDE-ALDH7A1 in existing biochemical NBS panels. Further prospective studies in larger cohorts are needed to refine cutoffs and confirm clinical performance. Full article
Show Figures

Figure 1

50 pages, 937 KiB  
Review
Precision Neuro-Oncology in Glioblastoma: AI-Guided CRISPR Editing and Real-Time Multi-Omics for Genomic Brain Surgery
by Matei Șerban, Corneliu Toader and Răzvan-Adrian Covache-Busuioc
Int. J. Mol. Sci. 2025, 26(15), 7364; https://doi.org/10.3390/ijms26157364 - 30 Jul 2025
Viewed by 413
Abstract
Precision neurosurgery is rapidly evolving as a medical specialty by merging genomic medicine, multi-omics technologies, and artificial intelligence (AI) technology, while at the same time, society is shifting away from the traditional, anatomic model of care to consider a more precise, molecular model [...] Read more.
Precision neurosurgery is rapidly evolving as a medical specialty by merging genomic medicine, multi-omics technologies, and artificial intelligence (AI) technology, while at the same time, society is shifting away from the traditional, anatomic model of care to consider a more precise, molecular model of care. The general purpose of this review is to contemporaneously reflect on how these advances will impact neurosurgical care by providing us with more precise diagnostic and treatment pathways. We hope to provide a relevant review of the recent advances in genomics and multi-omics in the context of clinical practice and highlight their transformational opportunities in the existing models of care, where improved molecular insights can support improvements in clinical care. More specifically, we will highlight how genomic profiling, CRISPR-Cas9, and multi-omics platforms (genomics, transcriptomics, proteomics, and metabolomics) are increasing our understanding of central nervous system (CNS) disorders. Achievements obtained with transformational technologies such as single-cell RNA sequencing and intraoperative mass spectrometry are exemplary of the molecular diagnostic possibilities in real-time molecular diagnostics to enable a more directed approach in surgical options. We will also explore how identifying specific biomarkers (e.g., IDH mutations and MGMT promoter methylation) became a tipping point in the care of glioblastoma and allowed for the establishment of a new taxonomy of tumors that became applicable for surgeons, where a change in practice enjoined a different surgical resection approach and subsequently stratified the adjuvant therapies undertaken after surgery. Furthermore, we reflect on how the novel genomic characterization of mutations like DEPDC5 and SCN1A transformed the pre-surgery selection of surgical candidates for refractory epilepsy when conventional imaging did not define an epileptogenic zone, thus reducing resective surgery occurring in clinical practice. While we are atop the crest of an exciting wave of advances, we recognize that we also must be diligent about the challenges we must navigate to implement genomic medicine in neurosurgery—including ethical and technical challenges that could arise when genomic mutation-based therapies require the concurrent application of multi-omics data collection to be realized in practice for the benefit of patients, as well as the constraints from the blood–brain barrier. The primary challenges also relate to the possible gene privacy implications around genomic medicine and equitable access to technology-based alternative practice disrupting interventions. We hope the contribution from this review will not just be situational consolidation and integration of knowledge but also a stimulus for new lines of research and clinical practice. We also hope to stimulate mindful discussions about future possibilities for conscientious and sustainable progress in our evolution toward a genomic model of precision neurosurgery. In the spirit of providing a critical perspective, we hope that we are also adding to the larger opportunity to embed molecular precision into neuroscience care, striving to promote better practice and better outcomes for patients in a global sense. Full article
(This article belongs to the Special Issue Molecular Insights into Glioblastoma Pathogenesis and Therapeutics)
Show Figures

Figure 1

16 pages, 745 KiB  
Article
Immature Platelet Fraction as a Sensitive Biomarker in Neonatal Sepsis: Diagnostic Performance Preceding Thrombocytopenia
by Ilkay Er and Medeni Arpa
Children 2025, 12(7), 931; https://doi.org/10.3390/children12070931 - 15 Jul 2025
Viewed by 243
Abstract
Background: Early and accurate diagnosis of neonatal sepsis remains a clinical challenge due to nonspecific signs and limitations of conventional biomarkers. The immature platelet fraction (IPF), a novel hematologic parameter reflecting thrombopoietic activity, has emerged as a potential early sepsis indicator. This [...] Read more.
Background: Early and accurate diagnosis of neonatal sepsis remains a clinical challenge due to nonspecific signs and limitations of conventional biomarkers. The immature platelet fraction (IPF), a novel hematologic parameter reflecting thrombopoietic activity, has emerged as a potential early sepsis indicator. This study aimed to evaluate the diagnostic value of IPF in neonatal sepsis prior to the onset of thrombocytopenia. Methods: This prospective study enrolled neonates with early-onset sepsis (EOS), late-onset sepsis (LOS), and healthy controls. IPF, C-reactive protein (CRP), procalcitonin (PCT), and hematologic indices were measured at diagnosis and 48–72 h post-treatment. Diagnostic performance was evaluated via ROC curve analysis, and correlations between IPF and inflammatory/hematologic markers were examined. IPF levels were also compared based on blood culture results. Results: IPF levels were significantly higher in both EOS (n: 56) and LOS (n: 50) groups compared to controls (n: 44) (p < 0.001). ROC analysis showed excellent diagnostic performance, with AUCs of 0.98 (EOS) and 0.99 (LOS). Following antibiotic treatment, IPF levels declined significantly (p < 0.001), supporting its dynamic value. Strong and moderate correlations were found with MPV and CRP, respectively, and an inverse association with platelet count, but not with PCT. Moreover, IPF levels were higher in culture-positive cases compared to culture-negative ones (13.1% vs. 9.8%; p = 0.017) and exhibited diagnostic performance comparable to CRP in predicting blood culture positivity. Conclusions: This study presents original and clinically relevant data supporting IPF as a promising and practical hematologic biomarker for early detection and treatment monitoring of neonatal sepsis. Its integration into standard sepsis evaluation protocols may improve early risk stratification and clinical decision-making in neonatal intensive care settings. Full article
(This article belongs to the Special Issue Providing Care for Preterm Infants)
Show Figures

Figure 1

9 pages, 393 KiB  
Article
TARE-Induced Pan-Immune Inflammation Value as a Prognostic Biomarker in Liver-Dominant Metastatic Colorectal Cancer
by Bengu Dursun, Burak Demir, Nejat Emre Öksüz, Çiğdem Soydal and Güngör Utkan
J. Clin. Med. 2025, 14(14), 4927; https://doi.org/10.3390/jcm14144927 - 11 Jul 2025
Viewed by 385
Abstract
Purpose: Previous studies have reported that blood-based inflammatory markers are associated with prognosis in patients with various solid tumors, including colorectal cancer (CRC). The pan-immune inflammation value (PIV) is a novel prognostic biomarker based on blood count. Here, we aimed to study the [...] Read more.
Purpose: Previous studies have reported that blood-based inflammatory markers are associated with prognosis in patients with various solid tumors, including colorectal cancer (CRC). The pan-immune inflammation value (PIV) is a novel prognostic biomarker based on blood count. Here, we aimed to study the association between PIV and survival following transarterial radioembolization (TARE) in patients with liver-dominant metastatic colorectal cancer (CLM). Methods: A total of 49 patients with CLM who underwent TARE at the Ankara University Department of Medical Oncology were retrospectively analyzed. The relationship between clinical and laboratory parameters with post-TARE overall survival (OS) was analyzed by multivariate analyses. Results: The median age was 60 years and 71.4% of patients had received at least two lines of chemotherapy. The objective response rate (ORR) was 59.1% following TARE. Patients with hepatic response after TARE treatment demonstrated significantly longer survival compared to non-responders (p: 0.033). The optimal PIV threshold value was calculated as 629 in ROC analyses. This PIV value had 81% sensitivity and 80% specificity for OS prediction (AUC 0.86; 95% CI: 0.75–0.98, p = 0.008). Patients with elevated PIV > 629 had significantly shorter OS (p = 0.002). In the multivariate analysis, adjusted for ECOG PS, TARE response, presence of extrahepatic disease, number of chemotherapy lines, CEA levels and post-TARE NLR and PIV, only low PIV level was associated with longer OS (>629 vs. ≤629; HR: 4.87; 95% CI: 1.32–17.92; p = 0.017). Conclusions: PIV, a blood-based inflammatory score, may reflect the host’s immune response following TARE and serve as a novel predictor of survival. Full article
(This article belongs to the Section Oncology)
Show Figures

Figure 1

18 pages, 1436 KiB  
Article
Circulating Bacterial DNA as a Novel Blood-Based Biomarker in Type 2 Diabetes Mellitus (DM2): Results from the PROMOTERA Study
by Robertina Giacconi, Patrizia D’Aquila, Fabiola Olivieri, Davide Gentilini, Luciano Calzari, Carlo Fortunato, Gretta Veronica Badillo Pazmay, Mirko Di Rosa, Giada Sena, Elisabetta De Rose, Antonio Cherubini, Riccardo Sarzani, Roberto Antonicelli, Giuseppe Pelliccioni, Anna Rita Bonfigli, Roberta Galeazzi, Fabrizia Lattanzio, Giuseppe Passarino, Dina Bellizzi and Francesco Piacenza
Int. J. Mol. Sci. 2025, 26(14), 6564; https://doi.org/10.3390/ijms26146564 - 8 Jul 2025
Viewed by 408
Abstract
Blood bacterial DNA (BB-DNA) has been identified as a novel biomarker for metabolic dysfunction, yet its relationship with epigenetic features in type 2 diabetes mellitus (DM2) patients remains largely unexplored. This study investigated the relationship between BB-DNA and epigenetic, inflammatory, and aging-related markers [...] Read more.
Blood bacterial DNA (BB-DNA) has been identified as a novel biomarker for metabolic dysfunction, yet its relationship with epigenetic features in type 2 diabetes mellitus (DM2) patients remains largely unexplored. This study investigated the relationship between BB-DNA and epigenetic, inflammatory, and aging-related markers in 285 elderly both with and without DM2. BB-DNA levels were higher in DM2 patients than in non-diabetic subjects, with the highest levels in those with severe renal impairment. BB-DNA showed a positive association with plasma IL-1β, linking bacterial DNA to systemic inflammation. Epigenetic analysis revealed a negative correlation between BB-DNA and DNA methylation-based leukocyte telomere length, suggesting accelerated aging in DM2. Additionally, BB-DNA was positively associated with DNAm-based biological age estimators, particularly DNAmPhenoAge and DNAmAge Skin Blood Clock. BB-DNA also correlated with DNAmVEGFA and DNAmCystatin C, key markers of diabetic nephropathy and vascular dysfunction. Furthermore, BB-DNA levels were associated with hypomethylation of genes involved in inflammation (e.g., IL1β, TNFα, IFNγ), cellular senescence (p16, p21, TP53), and metabolic regulation (e.g., IGF1, SREBF1, ABCG1, PDK4). These associations suggest that increased BB-DNA may reflect and potentially promote a pro-inflammatory and pro-senescent epigenetic profile in DM2. Importantly, many of these associations remained significant after adjusting for diabetes status, supporting BB-DNA as a robust biomarker across clinical subgroups. These findings provide new insights into the relationship between BB-DNA, inflammation, and epigenetic aging in DM2, highlighting BB-DNA as a potential biomarker for disease progression and complications, particularly in relation to renal dysfunction and systemic inflammation. Full article
(This article belongs to the Section Molecular Endocrinology and Metabolism)
Show Figures

Figure 1

19 pages, 1766 KiB  
Review
A Critical Role of Neutrophil-Driven Amplification of Chronic Microinflammation in the Biocompatibility of Hemodialysis
by Masaaki Nakayama, Hiroyuki Miyakawa, Kazuya Ohama and Hirokazu Kimura
Int. J. Mol. Sci. 2025, 26(13), 6472; https://doi.org/10.3390/ijms26136472 - 4 Jul 2025
Viewed by 361
Abstract
This review highlights recent insights into the pathophysiology and therapeutic strategies for improving biocompatibility in hemodialysis. Hemodialysis activates the innate immune system, particularly the complement cascade and neutrophils, leading to acute microinflammation. Interleukin-8 (IL-8), which increases during dialysis, promotes neutrophil chemotaxis and neutrophil [...] Read more.
This review highlights recent insights into the pathophysiology and therapeutic strategies for improving biocompatibility in hemodialysis. Hemodialysis activates the innate immune system, particularly the complement cascade and neutrophils, leading to acute microinflammation. Interleukin-8 (IL-8), which increases during dialysis, promotes neutrophil chemotaxis and neutrophil extracellular trap (NET) formation, triggering myeloperoxidase (MPO) release and oxidative stress. Neutrophil accumulation in atherosclerotic plaques exacerbates vascular inflammation through IL-6 upregulation. Elevated levels of IL-8, MPO, and NET-related biomarkers are associated with increased all-cause and cardiovascular mortality in dialysis patients. Strategies to mitigate these effects include the use of advanced membrane materials (e.g., AN69, vitamin E-coated, polymethyl methacrylate), novel dialysis modalities (e.g., high-volume online hemodiafiltration, cool dialysate, hydrogen-enriched dialysate), and citrate-based anticoagulation. These approaches aim to suppress complement activation, reduce oxidative stress, and limit neutrophil-induced damage. Enhancing biocompatibility is crucial for reducing cardiovascular complications and improving outcomes in dialysis patients. Suppressing the innate immune response during dialysis may become a future cornerstone in extracorporeal blood purification therapy. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

21 pages, 719 KiB  
Review
Biologic Therapy in Severe Asthma: A Phenotype-Driven and Targeted Approach
by Maria D’Amato, Daniela Pastore, Chiara Lupia, Claudio Candia, Andrea Bruni, Eugenio Garofalo, Federico Longhini, Angelantonio Maglio, Albino Petrone, Alessandro Vatrella, Girolamo Pelaia and Corrado Pelaia
J. Clin. Med. 2025, 14(13), 4749; https://doi.org/10.3390/jcm14134749 - 4 Jul 2025
Viewed by 963
Abstract
Asthma is a highly heterogeneous respiratory disease that, in its severe forms, is characterized by persistent symptoms, frequent exacerbations, and a significant impact on patients’ quality of life. Despite high-dose inhaled corticosteroids and long-acting bronchodilators, a subset of patients remains uncontrolled, necessitating advanced [...] Read more.
Asthma is a highly heterogeneous respiratory disease that, in its severe forms, is characterized by persistent symptoms, frequent exacerbations, and a significant impact on patients’ quality of life. Despite high-dose inhaled corticosteroids and long-acting bronchodilators, a subset of patients remains uncontrolled, necessitating advanced therapeutic strategies. The advent of biologic therapies has revolutionized the management of severe asthma, offering targeted interventions based on the underlying inflammatory endotypes, primarily T2-high and T2-low. However, selecting the most appropriate biologic remains challenging due to overlapping phenotypic features and the limited availability of validated biomarkers. This narrative review explores the clinical utility of key biomarkers, including blood eosinophils, fractional exhaled nitric oxide (FeNO), periostin, and total and specific IgE, in guiding biologic therapy. All the information provided is based on an extensive literature search conducted on PubMed. We also examine the clinical characteristics and comorbidities that influence therapeutic choices. Furthermore, we present a practical decision-making platform, including a clinical table matching phenotypes with biologic agents, such as omalizumab, mepolizumab, benralizumab, dupilumab, and tezepelumab. By integrating biomarker analysis with clinical assessment, based on current guidelines and our extensive real-life experience, we aim to offer a logical framework to help clinicians select the most suitable biologic treatment for patients with uncontrolled severe asthma. Future research should focus on identifying novel biomarkers, refining patient stratification, and evaluating long-term outcomes to further advance precision medicine in the management of severe asthma. Full article
(This article belongs to the Special Issue New Clinical Advances in Chronic Asthma)
Show Figures

Figure 1

28 pages, 5449 KiB  
Article
The Impact of Peroxiredoxin 3 on Molecular Testing, Diagnosis, and Prognosis in Human Pancreatic Ductal Adenocarcinoma
by Anna Kakehashi, Shugo Suzuki, Yusaku Nishidoi, Atsushi Hagihara, Hiroko Ikenaga, Masayuki Shiota, Guiyu Qiu, Ikue Noura, Yuko Kuwae, Arpamas Vachiraarunwong, Masaki Fujioka, Min Gi, Norifumi Kawada and Hideki Wanibuchi
Cancers 2025, 17(13), 2212; https://doi.org/10.3390/cancers17132212 - 1 Jul 2025
Viewed by 464
Abstract
Background/Objective: Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer death and tumors with an extremely poor prognosis. In the present study, novel biomarker candidates useful for the early diagnosis and prognosis of human invasive PDAC were investigated. Methods: Biomarker [...] Read more.
Background/Objective: Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer death and tumors with an extremely poor prognosis. In the present study, novel biomarker candidates useful for the early diagnosis and prognosis of human invasive PDAC were investigated. Methods: Biomarker candidates were first selected based on the proteomic/bioinformatic and clinico-pathological analyses of 10 and 100 patients with PDAC, respectively, operated at Osaka Metropolitan University Hospital (Exp. 1). Next, the expression and secretion of the target protein and its EV mRNA were investigated in pancreatic cancer cells in vitro and in a Balb/c nude mouse model. In addition, the protein and EV mRNA levels of candidate molecules were measured in the blood serum of 36 PDAC and 10 IPMN patients, and diagnostic significance was assessed (Exp. 2). Results: A significant elevation of peroxiredoxin 3 (PRX3), a mitochondrial matrix protein, was found in PDAC via LC-Ms/Ms analysis. In Exp. 1, PRX3 overexpression was found in PDAC and PanIN lesions and was associated with a tumor infiltrative growth pattern (INFc) and poor overall 1-year patient survival. The prognostic value was significantly improved when PRX3 was combined with serum SPan-1 and DUPAN-2 markers in survival analyses. Furthermore, the PRX3 protein and its extracellular vesicle (EV: exosome and oncosome)-incorporated mRNA were secreted at detectable levels from PANC-1, MIAPaCa-2, and SW1990 cells into the blood of Balb/c nude mice bearing tumors. The overexpression of PRX3 was positively correlated with that of cancer stem cell marker CD44 variant 9 (CD44v9), P-Nrf2, and FOXO3a, as well as the generation of reactive oxygen species. In Exp. 2, a significant increase in PRX3 protein and EV mRNA was detected in the blood serum of PDAC subjects compared to IPMN patients and healthy controls. Significantly higher PRX3 protein levels were found in the IPMN group. The elevation of PRX3 EV mRNA was significantly associated with poor patient survival. Conclusions: These results indicate that PRX3 may become a novel early biomarker for PDAC diagnosis and prognosis. Full article
Show Figures

Figure 1

31 pages, 1395 KiB  
Review
Salivary Interleukins as Non-Invasive Biomarkers for Psoriasis: Advances and Challenges in Diagnosis and Monitoring
by Anna Sora, Tony Hangan, Sergiu Ioachim Chirila, Leonard Gurgas, Mihaela Botnarciuc, Lavinia Carmen Daba, Ana Maria Cretu, Ionut Burlacu, Mihaela Zamfirescu, Adina Petcu, Adrian Cosmin Rosca, Ramona Mihaela Stoicescu and Lucian Cristian Petcu
Medicina 2025, 61(7), 1180; https://doi.org/10.3390/medicina61071180 - 29 Jun 2025
Viewed by 623
Abstract
Background and Objectives: Psoriasis is a chronic immune-mediated inflammatory disease requiring reliable diagnostic and monitoring tools. Salivary interleukins have emerged as promising non-invasive biomarkers, reflecting systemic inflammation and offering practical advantages such as ease of collection and improved patient compliance. Materials and [...] Read more.
Background and Objectives: Psoriasis is a chronic immune-mediated inflammatory disease requiring reliable diagnostic and monitoring tools. Salivary interleukins have emerged as promising non-invasive biomarkers, reflecting systemic inflammation and offering practical advantages such as ease of collection and improved patient compliance. Materials and Methods: This review synthesizes the current evidence on key salivary cytokines—IL-1β, IL-6, TNF-α, and IL-17—in relation to psoriasis pathogenesis, diagnosis, and treatment monitoring. It also compares saliva to blood-based diagnostics, emphasizing benefits like cost-effectiveness and suitability for repeated sampling. Methodological challenges, including heterogeneity in collection protocols and limited longitudinal data, are critically examined. Results: Advances in biologic therapies have deepened the understanding of psoriasis immunopathogenesis, highlighting interleukins as central biomarkers. Recent findings identify IL-37 and IL-38 as novel regulatory cytokines with anti-inflammatory roles. While elevated serum TNF-α levels in psoriatic patients are well documented, some inconsistencies persist. Notably, saliva has proven to be a viable alternative diagnostic fluid, supporting large-scale screening and routine clinical monitoring. Conclusions: Salivary interleukins—particularly IL-1β, IL-6, TNF-α, and IL-17—represent valuable, non-invasive biomarkers for early detection, disease severity assessment, and therapeutic response monitoring in psoriasis. Standardizing saliva-based methods and conducting large-scale studies are essential next steps to support their integration into personalized clinical practice. Full article
Show Figures

Figure 1

24 pages, 7389 KiB  
Article
A Novel Approach to Retinal Blood Vessel Segmentation Using Bi-LSTM-Based Networks
by Pere Marti-Puig, Kevin Mamaqi Kapllani and Bartomeu Ayala-Márquez
Mathematics 2025, 13(13), 2043; https://doi.org/10.3390/math13132043 - 20 Jun 2025
Viewed by 472
Abstract
The morphology of blood vessels in retinal fundus images is a key biomarker for diagnosing conditions such as glaucoma, hypertension, and diabetic retinopathy. This study introduces a deep learning-based method for automatic blood vessel segmentation, trained from scratch on 44 clinician-annotated images. The [...] Read more.
The morphology of blood vessels in retinal fundus images is a key biomarker for diagnosing conditions such as glaucoma, hypertension, and diabetic retinopathy. This study introduces a deep learning-based method for automatic blood vessel segmentation, trained from scratch on 44 clinician-annotated images. The proposed architecture integrates Bidirectional Long Short-Term Memory (Bi-LSTM) layers with dropout to mitigate overfitting. A distinguishing feature of this approach is the column-wise processing, which improves feature extraction and segmentation accuracy. Additionally, a custom data augmentation technique tailored for retinal images is implemented to improve training performance. The results are presented in their raw form—without post-processing—to objectively assess the method’s effectiveness and limitations. Further refinements, including pre- and post-processing and the use of image rotations to combine multiple segmentation outputs, could significantly boost performance. Overall, this work offers a novel and effective approach to the still unresolved task of retinal vessel segmentation, contributing to more reliable automated analysis in ophthalmic diagnostics. Full article
(This article belongs to the Special Issue Intelligent Computing with Applications in Computer Vision)
Show Figures

Figure 1

17 pages, 2159 KiB  
Article
Feasibility of Multiplex Cytokine Profiling in Preterm Labor: Towards Biomarker Discovery
by Ruth Llano, Inés Ardao, José Manuel Brea, Luz Romero, María P. Pata, Antón L. Martínez, Manuel Macía and María Isabel Loza
Biology 2025, 14(6), 714; https://doi.org/10.3390/biology14060714 - 17 Jun 2025
Viewed by 436
Abstract
Preterm delivery affects approximately 10% of pregnancies worldwide and remains a major clinical challenge due to the lack of reliable early predictive tools. Existing strategies are often invasive, relying on blood or amniotic fluid samples and requiring complex processing. In this study, we [...] Read more.
Preterm delivery affects approximately 10% of pregnancies worldwide and remains a major clinical challenge due to the lack of reliable early predictive tools. Existing strategies are often invasive, relying on blood or amniotic fluid samples and requiring complex processing. In this study, we describe a novel non-invasive approach based on the multiplex detection of inflammatory cytokines in small urine volumes from pregnant women. To account for clinical and temporal variability, we applied Generalized Additive Models for Location, Scale, and Shape (GAMLSS) to adjust for gestational age at sampling and obstetric factors. Correlation network analyses revealed cytokine interactions that distinguished preterm from term deliveries, with macrophage-derived cytokines—MIP-1α, MIP-1β, IL-15, and IL-22—emerging as central nodes. These findings highlight the involvement of the IL-1 pathway in the pathophysiology of preterm labor. Furthermore, urinary IL-5 and IL-31 levels correlated positively with pregnancy duration, whereas IL-1β and IL-1Ra in urine and TNFα in amniotic fluid showed inverse associations. Altogether, this non-invasive methodology provides insight into immune dynamics during pregnancy and offers a foundation for future studies focused on biomarker discovery and mechanistic understanding of preterm birth. Full article
Show Figures

Graphical abstract

35 pages, 1619 KiB  
Review
Blood-Based Biomarkers as Predictive and Prognostic Factors in Immunotherapy-Treated Patients with Solid Tumors—Currents and Perspectives
by Franciszek Kaczmarek, Anna Marcinkowska-Gapińska, Joanna Bartkowiak-Wieczorek, Michał Nowak, Michał Kmiecik, Kinga Brzezińska, Mariusz Dotka, Paweł Brosz, Wojciech Firlej and Paulina Wojtyła-Buciora
Cancers 2025, 17(12), 2001; https://doi.org/10.3390/cancers17122001 - 16 Jun 2025
Viewed by 1069
Abstract
Immunotherapy has revolutionized cancer treatment; however, the availability of cost-effective blood-based biomarkers for prognostic and predictive factors of immune treatment in patients with solid tumors remains limited. Due to low cost and easy accessibility, blood-based biomarkers should constitute an essential component of studies [...] Read more.
Immunotherapy has revolutionized cancer treatment; however, the availability of cost-effective blood-based biomarkers for prognostic and predictive factors of immune treatment in patients with solid tumors remains limited. Due to low cost and easy accessibility, blood-based biomarkers should constitute an essential component of studies to optimize and monitor immunotherapy. Currently available markers that can be measured in peripheral blood include total monocyte count, myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs), relative eosinophil count, cytokine levels (such as IL-6, IL-8, and IL-10), lactate dehydrogenase (LDH), C-reactive protein (CRP), soluble forms of CTLA-4 and PD-1 or PD-L1, as well as circulating tumor DNA (ctDNA). In our mini-review, we discuss the latest evidence indicating that routinely accessible peripheral blood parameters—such as the neutrophil-to-lymphocyte ratio (NLR), lymphocyte-to-monocyte ratio (LMR), platelet-to-lymphocyte ratio (PLR), and rheological parameters, which so far have been rarely considered for such an application, may be used as non-invasive biomarkers in cancer immunotherapy. Rheological parameters such as whole blood viscosity are influenced by several factors, such as hematocrit, aggregability and deformability of erythrocytes, and plasma viscosity, which is largely dependent on plasma proteins. Especially in cases where the set of symptoms indicates a high probability of hyperviscosity syndrome, blood rheological tests can lead to early diagnosis and treatment. Both biochemical and rheological parameters are prone to become novel and future standards for assessing immunotherapy among patients with solid tumors. Full article
Show Figures

Graphical abstract

30 pages, 1043 KiB  
Review
Perspectives in Amyotrophic Lateral Sclerosis: Biomarkers, Omics, and Gene Therapy Informing Disease and Treatment
by Nina Bono, Flaminia Fruzzetti, Giorgia Farinazzo, Gabriele Candiani and Stefania Marcuzzo
Int. J. Mol. Sci. 2025, 26(12), 5671; https://doi.org/10.3390/ijms26125671 - 13 Jun 2025
Viewed by 1588
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive loss of upper and lower motor neurons, leading to muscle weakness, paralysis, and ultimately respiratory failure. Despite advances in understanding its genetic basis, particularly mutations in Chromosome 9 Open Reading [...] Read more.
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive loss of upper and lower motor neurons, leading to muscle weakness, paralysis, and ultimately respiratory failure. Despite advances in understanding its genetic basis, particularly mutations in Chromosome 9 Open Reading Frame 72 (C9orf72), superoxide dismutase 1 (SOD1), TAR DNA-binding protein (TARDBP), and Fused in Sarcoma (FUS) gene, current diagnostic methods result in delayed intervention, and available treatments offer only modest benefits. This review examines innovative approaches transforming ALS research and clinical management. We explore emerging biomarkers, including the fluid-based markers such as neurofilament light chain, exosomes, and microRNAs in biological fluids, alongside the non-fluid-based biomarkers, including neuroimaging and electrophysiological markers, for early diagnosis and patient stratification. The integration of multi-omics data reveals complex molecular mechanisms underlying ALS heterogeneity, potentially identifying novel therapeutic targets. We highlight current gene therapy strategies, including antisense oligonucleotides (ASOs), RNA interference (RNAi), and CRISPR/Cas9 gene editing systems, alongside advanced delivery methods for crossing the blood–brain barrier. By bridging molecular neuroscience with bioengineering, these technologies promise to revolutionize ALS diagnosis and treatment, advancing toward truly disease-modifying interventions for this previously intractable condition. Full article
(This article belongs to the Special Issue Amyotrophic Lateral Sclerosis (ALS): Pathogenesis and Treatments)
Show Figures

Figure 1

47 pages, 2976 KiB  
Review
Epigenetic Alterations in Glioblastoma Multiforme as Novel Therapeutic Targets: A Scoping Review
by Marco Meleiro and Rui Henrique
Int. J. Mol. Sci. 2025, 26(12), 5634; https://doi.org/10.3390/ijms26125634 - 12 Jun 2025
Viewed by 1359
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive primary brain tumor with a dismal prognosis despite advances in multimodal treatment. Conventional therapies fail to achieve durable responses due to GBM’s molecular heterogeneity and capacity to evade therapeutic pressures. Epigenetic alterations have emerged as critical [...] Read more.
Glioblastoma multiforme (GBM) is a highly aggressive primary brain tumor with a dismal prognosis despite advances in multimodal treatment. Conventional therapies fail to achieve durable responses due to GBM’s molecular heterogeneity and capacity to evade therapeutic pressures. Epigenetic alterations have emerged as critical contributors to GBM pathobiology, including aberrant DNA methylation, histone modifications, and non-coding RNA (ncRNA) dysregulation. These mechanisms drive oncogenesis, therapy resistance, and immune evasion. This scoping review evaluates the current state of knowledge on epigenetic modifications in GBM, synthesizing findings from original articles and preclinical and clinical trials published over the last decade. Particular attention is given to MGMT promoter hypermethylation status as a biomarker for temozolomide (TMZ) sensitivity, histone deacetylation and methylation as modulators of chromatin structure, and microRNAs as regulators of pathways such as apoptosis and angiogenesis. Therapeutically, epigenetic drugs, like DNA methyltransferase inhibitors (DNMTis) and histone deacetylase inhibitors (HDACis), appear as promising approaches in preclinical models and early trials. Emerging RNA-based therapies targeting dysregulated ncRNAs represent a novel approach to reprogram the tumor epigenome. Combination therapies, pairing epigenetic agents with immune checkpoint inhibitors or chemotherapy, are explored for their potential to enhance treatment response. Despite these advancements, challenges such as tumor heterogeneity, the blood–brain barrier (BBB), and off-target effects remain significant. Future directions emphasize integrative omics approaches to identify patient-specific targets and refine therapies. This article thus highlights the potential of epigenetics in reshaping GBM treatment paradigms. Full article
(This article belongs to the Special Issue Glioblastoma: Molecular Pathogenesis and Treatment)
Show Figures

Graphical abstract

Back to TopTop