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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (224)

Search Parameters:
Keywords = cancer risk drivers

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 4023 KiB  
Article
Ferroptosis as a Therapeutic Avenue in Triple-Negative Breast Cancer: Mechanistic Insights and Prognostic Potential
by Taimoor Riaz, Muhammad Ali Saleem, Muhammad Umar Muzafar Khan, Muhammad Abdul Rehman Rashid and Muhammad Zubair
Biomedicines 2025, 13(8), 2037; https://doi.org/10.3390/biomedicines13082037 - 21 Aug 2025
Abstract
Background and Aims: Triple-negative breast cancer (TNBC) is a clinically aggressive malignancy marked by rapid disease progression, limited therapeutic avenues, and high recurrence risk. Ferroptosis an iron-dependent, lipid peroxidation-driven form of regulated cell death that has emerged as a promising therapeutic vulnerability in [...] Read more.
Background and Aims: Triple-negative breast cancer (TNBC) is a clinically aggressive malignancy marked by rapid disease progression, limited therapeutic avenues, and high recurrence risk. Ferroptosis an iron-dependent, lipid peroxidation-driven form of regulated cell death that has emerged as a promising therapeutic vulnerability in oncology. This study delineates the ferroptosis-associated molecular architecture of TNBC to identify key regulatory genes with prognostic and translational significance. Methods: Transcriptomic profiles from the GSE103091 dataset (130 TNBC and 30 normal breast tissue samples) were analyzed to identify ferroptosis-related differentially expressed genes (DEGs) using GEO2R. Protein–protein interaction (PPI) networks were constructed via STRING and GeneMANIA, with functional enrichment performed through Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Reactome analyses. Prognostic relevance was evaluated using GEPIA, BC-GenExMiner, and Kaplan–Meier Plotter survival analyses. Results: Six ferroptosis drivers (MAPK1, TLR4, IFNG, ATM, ULK2, and ATF3) and five suppressors (NFS1, GCLC, TP63, CD44, and SRC) were identified alongside HMOX1, a bifunctional regulator with context-dependent pro- and anti-ferroptotic activity. Enrichment analyses revealed significant associations with oxidative stress regulation, autophagy, immune modulation, and tumor progression pathways. Elevated IFNG expression was consistently linked to improve overall, disease-free, and distant metastasis-free survival, underscoring its dual function in antitumor immunity and ferroptosis sensitization. Conclusions: Ferroptosis represents a critical axis in TNBC pathophysiology, with IFNG emerging as both a prognostic biomarker and a viable therapeutic target. These insights provide a mechanistic foundation for integrating ferroptosis-inducing agents with immunotherapeutic modalities to enhance clinical outcomes and overcome therapeutic resistance in TNBC. Full article
Show Figures

Figure 1

26 pages, 4445 KiB  
Review
Unraveling the Converging Roles of ASC-Dependent Inflammasomes, Interleukin-1 Superfamily Members, Serum Amyloid A, and Non-Sterile Inflammation in Disease Pathology and Fibrosis in Inflammatory Bowel Disease and Primary Sclerosing Cholangitis
by Marco Losa, Marlene Schwarzfischer, Marc Emmenegger, Marianne R. Spalinger, Gerhard Rogler and Michael Scharl
Int. J. Mol. Sci. 2025, 26(16), 8042; https://doi.org/10.3390/ijms26168042 - 20 Aug 2025
Viewed by 256
Abstract
Inflammatory bowel disease (IBD) and primary sclerosing cholangitis (PSC) are chronic immune-mediated inflammatory diseases (IMIDs) that affect the gastrointestinal and hepatobiliary systems. They are characterized by persistent inflammation, potentially progressive fibrosis, and an elevated risk of developing cholangiocarcinoma and colorectal cancer. IBD and [...] Read more.
Inflammatory bowel disease (IBD) and primary sclerosing cholangitis (PSC) are chronic immune-mediated inflammatory diseases (IMIDs) that affect the gastrointestinal and hepatobiliary systems. They are characterized by persistent inflammation, potentially progressive fibrosis, and an elevated risk of developing cholangiocarcinoma and colorectal cancer. IBD and PSC share phenotypical, genetic, and immunological features, largely due to the central role of immune cell dysregulation. Despite their increasing global prevalence, the underlying drivers remain poorly understood, and effective treatment options are still lacking. Efforts towards an improved comprehension of their pathogenic mechanisms are therefore pivotal. Emerging evidence highlights the role of canonical ASC-dependent inflammasomes—multiprotein bioactive Interleukin (IL)-1-producing complexes of the innate immune system—and serum amyloid A (SAA) as key structures of gastrointestinal and hepatobiliary inflammation, tissue remodeling, stromal crosstalk, and fibrosis. In this review, we explore immunological connections and analogies between IBD and PSC, highlighting the converging roles of canonical ASC-dependent inflammasomes, the IL-1 superfamily, SAA, and sustained gut microbiota-driven chronic inflammation in disease pathology and their surging potential as therapeutic targets across the gut–liver axis. Full article
(This article belongs to the Special Issue Cellular and Molecular Mechanisms in Inflammatory Bowel Disease)
Show Figures

Figure 1

16 pages, 472 KiB  
Review
Beyond the Tissue: Unlocking NSCLC Treatment Potential Through Liquid Biopsy
by Milica Kontic, Mihailo Stjepanovic and Filip Markovic
Genes 2025, 16(8), 954; https://doi.org/10.3390/genes16080954 - 13 Aug 2025
Viewed by 409
Abstract
Lung cancer (LC), with non-small-cell lung cancer (NSCLC) as its predominant subtype, remains the leading cause of cancer-related mortality worldwide. While immune checkpoint inhibitors (ICIs) have redefined the therapeutic paradigm in advanced NSCLC, durable responses are confined to a limited subset of patients. [...] Read more.
Lung cancer (LC), with non-small-cell lung cancer (NSCLC) as its predominant subtype, remains the leading cause of cancer-related mortality worldwide. While immune checkpoint inhibitors (ICIs) have redefined the therapeutic paradigm in advanced NSCLC, durable responses are confined to a limited subset of patients. A major clinical challenge persists: the inability to accurately predict which patients will derive meaningful benefit, which will exhibit primary resistance, and which are at risk for severe immune-related toxicities. The imperative to individualize ICI therapy necessitates robust, dynamic, and accessible biomarkers. Liquid biopsy has emerged as a transformative, minimally invasive tool that enables real-time molecular and immunologic profiling. Through analysis of circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), exosomes, and peripheral blood immune components, liquid biopsy offers a window into both tumor intrinsic and host-related determinants of ICI response. These biomarkers not only hold promise for identifying predictive signatures—such as tumor mutational burden, neoantigen landscape, or immune activation states—but also for uncovering mechanisms of acquired resistance and guiding treatment adaptation. Beyond immunotherapy, liquid biopsy plays an increasingly central role in the landscape of targeted therapies, allowing early detection of actionable driver mutations and resistance mechanisms (e.g., EGFR T790M, MET amplification, and ALK fusion variants). Importantly, serial sampling via liquid biopsy facilitates longitudinal disease monitoring and timely therapeutic intervention without the need for repeated tissue biopsies. By guiding therapy selection, monitoring response, and detecting resistance early, liquid biopsy has the potential to significantly improve outcomes in NSCLC. Full article
(This article belongs to the Section Human Genomics and Genetic Diseases)
Show Figures

Figure 1

14 pages, 1672 KiB  
Article
Experiences of Advanced Non-Small Cell Lung Cancer Patients with Targeted Therapy Using Journey Mapping: A Qualitative Study
by Hailing Tu, Minghui Wang, Shengmei Yang and Jingfang Hong
Curr. Oncol. 2025, 32(8), 451; https://doi.org/10.3390/curroncol32080451 - 11 Aug 2025
Viewed by 425
Abstract
Targeted therapies against specific driver gene mutations have become the standard first-line treatment for most patients with advanced non-small cell lung cancer (NSCLC). While these therapies significantly prolong survival, the entire cancer treatment journey remains challenging and distressing. To better understand these experiences, [...] Read more.
Targeted therapies against specific driver gene mutations have become the standard first-line treatment for most patients with advanced non-small cell lung cancer (NSCLC). While these therapies significantly prolong survival, the entire cancer treatment journey remains challenging and distressing. To better understand these experiences, this study employed a qualitative descriptive approach, conducting semi-structured interviews with 18 advanced NSCLC patients receiving targeted therapy, supplemented by patient journey logs. The resulting journey map delineated five stages: diagnosis, initial treatment, maintenance therapy, disease progression, and end-of-life. The analysis identified four key themes characterizing patient experiences at each stage. These findings enable healthcare professionals to identify risk situations and determine optimal timing for support interventions. Similarly, preparing patients for the processes they must undergo and the side effects of medical treatment helps reduce their uncertainty and anxiety, thereby improving their quality of life. Full article
(This article belongs to the Section Thoracic Oncology)
Show Figures

Figure 1

33 pages, 1872 KiB  
Review
Exploring the Epidemiologic Burden, Pathogenetic Features, and Clinical Outcomes of Primary Liver Cancer in Patients with Type 2 Diabetes Mellitus (T2DM) and Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): A Scoping Review
by Mario Romeo, Fiammetta Di Nardo, Carmine Napolitano, Claudio Basile, Carlo Palma, Paolo Vaia, Marcello Dallio and Alessandro Federico
Diabetology 2025, 6(8), 79; https://doi.org/10.3390/diabetology6080079 - 4 Aug 2025
Viewed by 450
Abstract
Background/Objectives: Primary liver cancer (PLC), encompassing hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), constitutes a growing global health concern. Metabolic dysfunction-associated Steatotic Liver Disease (MASLD) and Type 2 diabetes mellitus (T2DM) represent a recurrent epidemiological overlap. Individuals with MASLD and T2DM (MASLD-T2DM) are [...] Read more.
Background/Objectives: Primary liver cancer (PLC), encompassing hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), constitutes a growing global health concern. Metabolic dysfunction-associated Steatotic Liver Disease (MASLD) and Type 2 diabetes mellitus (T2DM) represent a recurrent epidemiological overlap. Individuals with MASLD and T2DM (MASLD-T2DM) are at a higher risk of PLC. This scoping review highlights the epidemiological burden, the classic and novel pathogenetic frontiers, and the potential strategies optimizing the management of PLC in MASLD-T2DM. Methods: A systematic search of the PubMed, Medline, and SCOPUS electronic databases was conducted to identify evidence investigating the pathogenetic mechanisms linking MASLD and T2DM to hepatic carcinogenesis, highlighting the most relevant targets and the relatively emerging therapeutic strategies. The search algorithm included in sequence the filter words: “MASLD”, “liver steatosis”, “obesity”, “metabolic syndrome”, “body composition”, “insulin resistance”, “inflammation”, “oxidative stress”, “metabolic dysfunction”, “microbiota”, “glucose”, “immunometabolism”, “trained immunity”. Results: In the MASD-T2DM setting, insulin resistance (IR) and IR-induced mechanisms (including chronic inflammation, insulin/IGF-1 axis dysregulation, and autophagy), simultaneously with the alterations of gut microbiota composition and functioning, represent crucial pathogenetic factors in hepatocarcinogenesis. Besides, the glucose-related metabolic reprogramming emerged as a crucial pathogenetic moment contributing to cancer progression and immune evasion. In this scenario, lifestyle changes, simultaneously with antidiabetic drugs targeting IR-related effects and gut-liver axis, in parallel with novel approaches modulating immunometabolic pathways, represent promising strategies. Conclusions: Metabolic dysfunction, classically featuring MASLD-T2DM, constitutes a continuously expanding global issue, as well as a critical driver in PLC progression, demanding integrated and personalized interventions to reduce the future burden of disease. Full article
Show Figures

Figure 1

22 pages, 1013 KiB  
Review
Genomic Alterations and Microbiota Crosstalk in Hepatic Cancers: The Gut–Liver Axis in Tumorigenesis and Therapy
by Yuanji Fu, Jenny Bonifacio-Mundaca, Christophe Desterke, Íñigo Casafont and Jorge Mata-Garrido
Genes 2025, 16(8), 920; https://doi.org/10.3390/genes16080920 - 30 Jul 2025
Viewed by 392
Abstract
Background/Objectives: Hepatic cancers, including hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), are major global health concerns due to rising incidence and limited therapeutic success. While traditional risk factors include chronic liver disease and environmental exposures, recent evidence underscores the significance of genetic alterations and [...] Read more.
Background/Objectives: Hepatic cancers, including hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), are major global health concerns due to rising incidence and limited therapeutic success. While traditional risk factors include chronic liver disease and environmental exposures, recent evidence underscores the significance of genetic alterations and gut microbiota in liver cancer development and progression. This review aims to integrate emerging knowledge on the interplay between host genomic changes and gut microbial dynamics in the pathogenesis and treatment of hepatic cancers. Methods: We conducted a comprehensive review of current literature on genetic and epigenetic drivers of HCC and CCA, focusing on commonly mutated genes such as TP53, CTNNB1, TERT, IDH1/2, and FGFR2. In parallel, we evaluated studies addressing the gut–liver axis, including the roles of dysbiosis, microbial metabolites, and immune modulation. Key clinical and preclinical findings were synthesized to explore how host–microbe interactions influence tumorigenesis and therapeutic response. Results: HCC and CCA exhibit distinct but overlapping genomic landscapes marked by recurrent mutations and epigenetic reprogramming. Alterations in the gut microbiota contribute to hepatic inflammation, genomic instability, and immune evasion, potentially enhancing oncogenic signaling pathways. Furthermore, microbiota composition appears to affect responses to immune checkpoint inhibitors. Emerging therapeutic strategies such as probiotics, fecal microbiota transplantation, and precision oncology based on mutational profiling demonstrate potential for personalized interventions. Conclusions: The integration of host genomics with microbial ecology provides a promising paradigm for advancing diagnostics and therapies in liver cancer. Targeting the gut–liver axis may complement genome-informed strategies to improve outcomes for patients with HCC and CCA. Full article
(This article belongs to the Special Issue Feature Papers in Microbial Genetics and Genomics)
Show Figures

Figure 1

22 pages, 1317 KiB  
Review
Obesity: Clinical Impact, Pathophysiology, Complications, and Modern Innovations in Therapeutic Strategies
by Mohammad Iftekhar Ullah and Sadeka Tamanna
Medicines 2025, 12(3), 19; https://doi.org/10.3390/medicines12030019 - 28 Jul 2025
Viewed by 1319
Abstract
Obesity is a growing global health concern with widespread impacts on physical, psychological, and social well-being. Clinically, it is a major driver of type 2 diabetes (T2D), cardiovascular disease (CVD), non-alcoholic fatty liver disease (NAFLD), and cancer, reducing life expectancy by 5–20 years [...] Read more.
Obesity is a growing global health concern with widespread impacts on physical, psychological, and social well-being. Clinically, it is a major driver of type 2 diabetes (T2D), cardiovascular disease (CVD), non-alcoholic fatty liver disease (NAFLD), and cancer, reducing life expectancy by 5–20 years and imposing a staggering economic burden of USD 2 trillion annually (2.8% of global GDP). Despite its significant health and socioeconomic impact, earlier obesity medications, such as fenfluramine, sibutramine, and orlistat, fell short of expectations due to limited effectiveness, serious side effects including valvular heart disease and gastrointestinal issues, and high rates of treatment discontinuation. The advent of glucagon-like peptide-1 (GLP-1) receptor agonists (e.g., semaglutide, tirzepatide) has revolutionized obesity management. These agents demonstrate unprecedented efficacy, achieving 15–25% mean weight loss in clinical trials, alongside reducing major adverse cardiovascular events by 20% and T2D incidence by 72%. Emerging therapies, including oral GLP-1 agonists and triple-receptor agonists (e.g., retatrutide), promise enhanced tolerability and muscle preservation, potentially bridging the efficacy gap with bariatric surgery. However, challenges persist. High costs, supply shortages, and unequal access pose significant barriers to the widespread implementation of obesity treatment, particularly in low-resource settings. Gastrointestinal side effects and long-term safety concerns require close monitoring, while weight regain after medication discontinuation emphasizes the need for ongoing adherence and lifestyle support. This review highlights the transformative potential of incretin-based therapies while advocating for policy reforms to address cost barriers, equitable access, and preventive strategies. Future research must prioritize long-term cardiovascular outcome trials and mitigate emerging risks, such as sarcopenia and joint degeneration. A multidisciplinary approach combining pharmacotherapy, behavioral interventions, and systemic policy changes is critical to curbing the obesity epidemic and its downstream consequences. Full article
Show Figures

Figure 1

19 pages, 2337 KiB  
Article
Gas–Particle Partitioning and Temporal Dynamics of Pesticides in Urban Atmosphere Adjacent to Agriculture
by Dani Khoury, Supansa Chimjarn, Olivier Delhomme and Maurice Millet
Atmosphere 2025, 16(7), 873; https://doi.org/10.3390/atmos16070873 - 17 Jul 2025
Viewed by 312
Abstract
Air pollution caused by pesticide residues is an emerging concern in urban environments influenced by nearby agricultural activities. In this study, weekly air samples were collected between May 2018 and March 2020 in Strasbourg, France, to quantify 104 pesticides in both gas and [...] Read more.
Air pollution caused by pesticide residues is an emerging concern in urban environments influenced by nearby agricultural activities. In this study, weekly air samples were collected between May 2018 and March 2020 in Strasbourg, France, to quantify 104 pesticides in both gas and particle phases using GC-MS/MS and LC-MS/MS. Herbicides and fungicides were the most frequently detected classes, appearing in 98% of both phases followed by insecticides. Key compounds such as metalaxyl-M, diphenylamine, and bifenthrin were present in over 90% of samples. Concentrations ranged from 2.5 to 63 ng m−3 weekly, with cumulative annual loads exceeding 1200 ng m−3. Gas–particle partitioning revealed that highly volatile compounds like azinphos-ethyl favored the gas phase, while less volatile ones like bifenthrin and tebuconazole partitioned >95% into particles. A third-degree polynomial regression (R2 of 0.74) revealed a nonlinear relationship between Kₚ and particle-phase concentrations, highlighting a threshold above Kₚ of 0.025 beyond which compounds accumulate disproportionately in the particulate phase. Seasonal variability showed that 36% of the annual pesticide load occurred in autumn, with total airborne levels peaking near 400 ng m−3, while the lowest load occurred during summer. Principal component analysis identified rainfall and total suspended particles as major drivers of pesticide phase distribution. The inhalation health risk assessed yielded hazard index values < 1 × 10−7 for all population groups, suggesting negligible non-cancer risk. This study highlights the prevalence, seasonal dynamics, and partition behavior of airborne pesticides in urban air and underscores the need for regulatory attention to this overlooked exposure route. Full article
(This article belongs to the Section Air Quality)
Show Figures

Figure 1

22 pages, 3438 KiB  
Article
Revolutionizing Detection of Minimal Residual Disease in Breast Cancer Using Patient-Derived Gene Signature
by Chen Yeh, Hung-Chih Lai, Nathan Grabbe, Xavier Willett and Shu-Ti Lin
Onco 2025, 5(3), 35; https://doi.org/10.3390/onco5030035 - 12 Jul 2025
Viewed by 489
Abstract
Background: Many patients harbor minimal residual disease (MRD)—small clusters of residual tumor cells that survive therapy and evade conventional detection but drive recurrence. Although advances in molecular and computational methods have improved circulating tumor DNA (ctDNA)-based MRD detection, these approaches face challenges: ctDNA [...] Read more.
Background: Many patients harbor minimal residual disease (MRD)—small clusters of residual tumor cells that survive therapy and evade conventional detection but drive recurrence. Although advances in molecular and computational methods have improved circulating tumor DNA (ctDNA)-based MRD detection, these approaches face challenges: ctDNA shedding fluctuates widely across tumor types, disease stages, and histological features. Additionally, low levels of driver mutations originating from healthy tissues can create background noise, complicating the accurate identification of bona fide tumor-specific signals. These limitations underscore the need for refined technologies to further enhance MRD detection beyond DNA sequences in solid malignancies. Methods: Profiling circulating cell-free mRNA (cfmRNA), which is hyperactive in tumor and non-tumor microenvironments, could address these limitations to inform postoperative surveillance and treatment strategies. This study reported the development of OncoMRD BREAST, a customized, gene signature-informed cfmRNA assay for residual disease monitoring in breast cancer. OncoMRD BREAST introduces several advanced technologies that distinguish it from the existing ctDNA-MRD tests. It builds on the patient-derived gene signature for capturing tumor activities while introducing significant upgrades to its liquid biopsy transcriptomic profiling, digital scoring systems, and tracking capabilities. Results: The OncoMRD BREAST test processes inputs from multiple cutting-edge biomarkers—tumor and non-tumor microenvironment—to provide enhanced awareness of tumor activities in real time. By fusing data from these diverse intra- and inter-cellular networks, OncoMRD BREAST significantly improves the sensitivity and reliability of MRD detection and prognosis analysis, even under challenging and complex conditions. In a proof-of-concept real-world pilot trial, OncoMRD BREAST’s rapid quantification of potential tumor activity helped reduce the risk of incorrect treatment strategies, while advanced predictive analytics contributed to the overall benefits and improved outcomes of patients. Conclusions: By tailoring the assay to individual tumor profiles, we aimed to enhance early identification of residual disease and optimize therapeutic decision-making. OncoMRD BREAST is the world’s first and only gene signature-powered test for monitoring residual disease in solid tumors. Full article
Show Figures

Figure 1

26 pages, 6233 KiB  
Review
Colonic Aging and Colorectal Cancer: An Unignorable Interplay and Its Translational Implications
by Qiyan Yin, Fen Qin, Fangliu Gan, Guangxi Zhao, Ronghua Chen, Yue Wen, Xueyang Hua, Fugui Zeng, Yuezheng Zhang, Yuliang Xiao, Wenbing Xie and Yong Tao
Biology 2025, 14(7), 805; https://doi.org/10.3390/biology14070805 - 3 Jul 2025
Viewed by 697
Abstract
Colorectal cancer (CRC) incidence increases markedly with age, yet chronological age is an inadequate proxy for the complex biological processes involved. Colon aging, the intrinsic biological aging of the colonic tissue, is emerging as a crucial, active driver of CRC development. This review [...] Read more.
Colorectal cancer (CRC) incidence increases markedly with age, yet chronological age is an inadequate proxy for the complex biological processes involved. Colon aging, the intrinsic biological aging of the colonic tissue, is emerging as a crucial, active driver of CRC development. This review comprehensively analyzes the interplay between colon aging and CRC pathogenesis by examining fundamental hallmarks of aging—such as altered tissue homeostasis, epigenetic dysregulation, and microenvironmental shifts including chronic inflammation (inflammaging), gut microbiome dysbiosis, and extracellular matrix remodeling—manifest specifically within the aging colon to synergistically foster a pro-tumorigenic environment. Key findings synthesized from the literature highlight the critical roles of impaired colonic stem cell function, epithelial barrier disruption (“leaky gut”), persistent low-grade inflammation, and altered microbial communities and their metabolites in promoting CRC initiation and progression. Translating this mechanistic understanding holds significant promise: insights from colon aging research can inform novel biomarkers for improved early detection and risk stratification, guide the development of personalized preventative strategies and therapeutic interventions targeting aging pathways, and underpin public health initiatives focused on healthy colon aging. Ultimately, recognizing colon aging as a modifiable contributor to CRC offers a powerful avenue to potentially reduce CRC incidence and enhance patient outcomes, particularly in the vulnerable aging population. Full article
(This article belongs to the Section Cancer Biology)
Show Figures

Figure 1

21 pages, 1856 KiB  
Article
Decoding the CD36-Centric Axis in Gastric Cancer: Insights into Lipid Metabolism, Obesity, and Hypercholesterolemia
by Preyangsee Dutta, Dwaipayan Saha, Atanu Giri, Aseem Rai Bhatnagar and Abhijit Chakraborty
Int. J. Transl. Med. 2025, 5(3), 26; https://doi.org/10.3390/ijtm5030026 - 23 Jun 2025
Viewed by 930
Abstract
Background: Gastric cancer is a leading cause of cancer-related mortality worldwide, with approximately one million new cases diagnosed annually. While Helicobacter pylori infection remains a primary etiological factor, mounting evidence implicates obesity and lipid metabolic dysregulation, particularly in hypercholesterolemia, as emerging drivers of [...] Read more.
Background: Gastric cancer is a leading cause of cancer-related mortality worldwide, with approximately one million new cases diagnosed annually. While Helicobacter pylori infection remains a primary etiological factor, mounting evidence implicates obesity and lipid metabolic dysregulation, particularly in hypercholesterolemia, as emerging drivers of gastric tumorigenesis. This study investigates the molecular intersections between gastric cancer, obesity, and hypercholesterolemia through a comprehensive multi-omics and systems biology approach. Methods: We conducted integrative transcriptomic analysis of gastric adenocarcinoma using The Cancer Genome Atlas (TCGA) RNA-sequencing dataset (n = 623, 8863 genes), matched with standardized clinical metadata (n = 413). Differential gene expression between survival groups was assessed using Welch’s t-test with Benjamini–Hochberg correction (FDR < 0.05, |log2FC| ≥ 1). High-confidence gene sets for obesity (n = 128) and hypercholesterolemia (n = 97) were curated from the OMIM, STRING (confidence ≥ 0.7), and KEGG databases using hierarchical evidence-based prioritization. Overlapping gene signatures were identified, followed by pathway enrichment via Enrichr (KEGG 2021 Human) and protein–protein interaction (PPI) analysis using STRING v11.5 and Cytoscape v3.9.0. CD36’s prognostic value was evaluated via Kaplan–Meier and log-rank testing alongside clinicopathological correlations. Results: We identified 36 genes shared between obesity and gastric cancer, and 31 genes shared between hypercholesterolemia and gastric cancer. CD36 emerged as the only gene intersecting all three conditions, marking it as a unique molecular integrator. Enrichment analyses implicated dysregulated fatty acid uptake, adipocytokine signaling, cholesterol metabolism, and NF-κB-mediated inflammation as key pathways. Elevated CD36 expression was significantly correlated with higher tumor stage (p = 0.016), reduced overall survival (p = 0.001), and race-specific expression differences (p = 0.007). No sex-based differences in CD36 expression or survival were observed. Conclusions: CD36 is a central metabolic–oncogenic node linking obesity, hypercholesterolemia, and gastric cancer. It functions as both a mechanistic driver of tumor progression and a clinically actionable biomarker, particularly in metabolically comorbid patients. These findings provide a rationale for targeting CD36-driven pathways as part of a precision oncology strategy and highlight the need to incorporate metabolic profiling into gastric cancer risk assessment and treatment paradigms. Full article
Show Figures

Figure 1

25 pages, 540 KiB  
Review
Malignancies in Celiac Disease—A Hidden Threat with Diagnostic Pitfalls
by Aleksandra Kubas and Ewa Małecka-Wojciesko
Biomedicines 2025, 13(6), 1507; https://doi.org/10.3390/biomedicines13061507 - 19 Jun 2025
Viewed by 943
Abstract
Celiac disease (CeD) is an autoimmune disorder that is triggered by gluten ingestion in genetically predisposed individuals. Untreated or poorly controlled CeD leads to various disease complications, such as malnutrition, osteoporosis, autoimmune diseases, or refractory celiac disease (RCD). Accumulating recent research has highlighted [...] Read more.
Celiac disease (CeD) is an autoimmune disorder that is triggered by gluten ingestion in genetically predisposed individuals. Untreated or poorly controlled CeD leads to various disease complications, such as malnutrition, osteoporosis, autoimmune diseases, or refractory celiac disease (RCD). Accumulating recent research has highlighted the association between CeD and the development of malignancies, particularly enteropathy-associated T-cell lymphoma (EATL) and small bowel carcinoma (SBC), which are neoplasms with extremely poor prognoses. Genetic alterations in the JAK1–STAT3 pathway and the high prevalence of microsatellite instability may be the main drivers of CeD-associated lymphomagenesis and small bowel oncogenesis and therefore could be an attractive therapeutic target to block cancer transformation. However, to date, the risk factors and exact mechanisms underlying malignancy development in patients with CeD remain unclear, and prospective cohort studies that include molecular profiling are needed. Moreover, current guidelines on the management of CeD do not provide standardized protocols for cancer surveillance—particularly regarding screening intervals, risk stratification, and monitoring strategies for high-risk patients such as those with RCD. This paper reviews the existing knowledge on malignancies in CeD, highlights diagnostic challenges, and discusses future perspectives on the early detection, monitoring, and treatment of CeD-associated neoplasms. Full article
(This article belongs to the Section Cancer Biology and Oncology)
Show Figures

Figure 1

20 pages, 3969 KiB  
Article
Analysis of Novel DNA Adducts Derived from Acetaldehyde
by Yuuki Betsuyaku, Mina Motohashi, Akira Sassa, Takeji Takamura-Enya and Yukari Totsuka
Biomolecules 2025, 15(6), 878; https://doi.org/10.3390/biom15060878 - 16 Jun 2025
Viewed by 688
Abstract
Alcohol consumption is a known risk factor for esophageal and liver cancers. Recently, it was reported that mutation signatures characterized by T:A to C:G mutations (SBS16), which are suggested to be associated with alcohol intake, are frequently detected in esophageal, liver, and stomach [...] Read more.
Alcohol consumption is a known risk factor for esophageal and liver cancers. Recently, it was reported that mutation signatures characterized by T:A to C:G mutations (SBS16), which are suggested to be associated with alcohol intake, are frequently detected in esophageal, liver, and stomach cancers among the Japanese population. However, the scientific evidence linking alcohol consumption to SBS16 remains lacking. Acetaldehyde (AA), a carcinogenic metabolite of alcohol, is considered a key contributor to alcohol-related cancer development. Although the guanine adducts associated with alcohol exposure have been reported as part of its carcinogenic mechanism, an adenine adduct, N6-ethyl-deoxyadenosine (N6-ethyl-dA), a potential contributor to the SBS16 mutation pattern, was recently identified using a mass spectrometry-based DNA adductome approach. However, the mutagenicity assessment of N6-ethyl-dA using primer extension assays and the supF gene mutation test showed that this adenine adduct is not mutagenic. To identify another candidate as a driver adduct for SBS16, a DNA adductome approach was conducted, leading to the identification of a novel adenine adduct, 3-(2′-deoxyribos-1′-yl)-7,9-dimethyl-3,9-dihydro-7H-[1,3,5]oxadiazino[4,3-i]purine (N1-oxydiethylidene-dA), in which two AA molecules are bound to an adenine base. Moreover, N1-oxydiethylidene-dA was detected in mouse livers, and its levels increased following ethanol administration, suggesting that alcohol may contribute to SBS16 induction via the formation of N1-oxydiethylidene-dA. Full article
(This article belongs to the Special Issue Recent Advances in Adduct Science)
Show Figures

Figure 1

12 pages, 557 KiB  
Article
Clinician Recommendation for Hereditary Genetic Testing in Participants at Increased Risk for Hereditary Cancer
by Emerson Delacroix, Sarah Austin, John D. Rice, Elena Martinez Stoffel, Erika Koeppe, Jennifer J. Griggs and Ken Resnicow
Cancers 2025, 17(12), 1994; https://doi.org/10.3390/cancers17121994 - 14 Jun 2025
Viewed by 511
Abstract
Background: Despite clinical utility in managing hereditary cancers, genetic testing (GT) remains underutilized. While barriers include knowledge gaps and cost, clinician recommendation is a major driver of GT uptake, with rates varying by cancer type and family cancer history documentation. Methods: Adult participants [...] Read more.
Background: Despite clinical utility in managing hereditary cancers, genetic testing (GT) remains underutilized. While barriers include knowledge gaps and cost, clinician recommendation is a major driver of GT uptake, with rates varying by cancer type and family cancer history documentation. Methods: Adult participants (≥18 years) were recruited through multiple sources to complete a cancer family history survey for a larger intervention trial. Participants with personal or family history indicating increased hereditary cancer risk who had not undergone GT (N = 3001) were invited to complete a baseline survey. Multivariable logistic regression was used to analyze associations between demographics and cancer history by receipt of a clinician recommendation for GT. Results: Among 784 respondents, most were White (84.6%), female (58.4%), and over age 51 (75.3%), with 58.2% reporting a diagnosis of cancer. Only 14.0% reported receiving a clinician recommendation for GT, with lower recommendation rates among younger adults (20.1%), those reporting no financial stress (10.7%), and those with higher education (12.0%). Multivariate analysis showed participants who did not report financial stress (p = 0.049) were less likely to receive a recommendation. Discussion: These findings highlight disparities in GT recommendation by clinicians. Increased clinician education about indications for GT, the implementation of electronic medical record tools to facilitate the identification of patients with guideline-concordant personal and/or biological-relative cancer history, and patient-facing interventions could standardize the dissemination of recommendations for GT. Conclusions: Future efforts that focus on increasing clinician education and electronic decision support should identify individuals with personal and/or biological-relative cancer history meeting criteria for GT. Full article
(This article belongs to the Special Issue Socio-Demographic Factors and Cancer Research)
Show Figures

Figure 1

21 pages, 2183 KiB  
Article
Human Papillomavirus-Encoded microRNAs as Regulators of Human Gene Expression in Anal Squamous Cell Carcinoma: A Meta-Transcriptomics Study
by Daniel J. García, Marco A. Pulpillo-Berrocal, José L. Ruiz, Eduardo Andrés-León and Laura C. Terrón-Camero
Non-Coding RNA 2025, 11(3), 43; https://doi.org/10.3390/ncrna11030043 - 9 Jun 2025
Viewed by 839
Abstract
Introduction: Anal squamous cell carcinoma (ASCC) is a rare but increasingly common gastrointestinal malignancy, mainly associated with oncogenic human papillomaviruses (HPVs). The role of non-coding RNAs (ncRNAs) in tumorigenesis is recognized, but the impact of viral ncRNAs on host gene expression remains unclear. [...] Read more.
Introduction: Anal squamous cell carcinoma (ASCC) is a rare but increasingly common gastrointestinal malignancy, mainly associated with oncogenic human papillomaviruses (HPVs). The role of non-coding RNAs (ncRNAs) in tumorigenesis is recognized, but the impact of viral ncRNAs on host gene expression remains unclear. Methods: We re-analyzed total RNA-Seq data from 70 anal biopsies: 31 low-grade squamous intraepithelial lesions (LGSIL), 16 high-grade SIL (HGSIL), and 23 ASCC cases. Microbial composition was assessed taxonomically. Novel viral miRNAs were predicted using vsRNAfinder and linked to host targets using TargetScan and expression correlation analyses. Results: Microbial profiling revealed significant differences in abundance, with Alphapapillomaviruses types 9, 10, and 14 enriched across lesion grades. We identified 90 novel viral miRNAs and 177 significant anti-correlated miRNA–mRNA interactions. Target genes were enriched in pathways related to cell cycle, epithelial–mesenchymal transition, lipid metabolism, immune modulation, and viral replication. Discussion: Our findings suggest that HPV-derived miRNAs, including those from low-risk types, may contribute to neoplastic transformation by modulating host regulatory networks. Conclusion: This study highlights viral miRNAs as potential drivers of HPV-related anal cancer and supports their utility as early biomarkers and therapeutic targets in ASCC. Full article
Show Figures

Figure 1

Back to TopTop