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
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 (2,101)

Search Parameters:
Keywords = tumor fibroblasts

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
24 pages, 3579 KB  
Article
SIAH2–WNK1 Signaling Drives Glycolytic Metabolism and Therapeutic Resistance in Colorectal Cancer
by Kee-Thai Kiu, Cheng-Ying Chu, Yi-Chiao Cheng, Min-Hsuan Yen, Ying-Wei Chen, Narpati Wesa Pikatan, Vijesh Kumar Yadav and Tung-Cheng Chang
Int. J. Mol. Sci. 2026, 27(2), 1065; https://doi.org/10.3390/ijms27021065 - 21 Jan 2026
Abstract
Colorectal cancer (CRC) progression and therapy resistance are driven in part by metabolic reprogramming and the persistence of cancer stem-like cells (CSCs). The seven in absentia homolog 2 (SIAH2)/with-no-lysine kinase 1 (WNK1) signaling axis has emerged as a potential regulator of these processes, [...] Read more.
Colorectal cancer (CRC) progression and therapy resistance are driven in part by metabolic reprogramming and the persistence of cancer stem-like cells (CSCs). The seven in absentia homolog 2 (SIAH2)/with-no-lysine kinase 1 (WNK1) signaling axis has emerged as a potential regulator of these processes, yet its functional role in CRC metabolism and tumor–stroma crosstalk remains incompletely understood. Integrated analyses of The Cancer Genome Atlas–Colon Adenocarcinoma (TCGA-COAD) and Gene Expression Omnibus (GEO, GSE17538) datasets revealed significant upregulation of SIAH2 and WNK1 in CRC tissues, with strong positive correlations to glycolysis- and hypoxia-associated genes, including PFKP, LDHA, BPGM, ADH1A, ADH1B, and HIF-1α. Single-cell and clinical profiling further demonstrated preferential enrichment of SIAH2 in undifferentiated, stem-like tumor cell populations. Functional studies across multiple CRC cell lines showed that SIAH2 silencing suppressed proliferation, clonogenic growth, tumor sphere formation, and cell-cycle progression, whereas SIAH2 overexpression exerted opposite effects. Seahorse extracellular flux analyses established that SIAH2 promotes glycolytic capacity and metabolic flexibility. At the protein level, SIAH2 regulated glycolytic enzymes and WNK1/hypoxia-inducible factor-1α (HIF-1α) signaling, effects that were amplified by cancer-associated fibroblast (CAF)-derived conditioned medium. CAF exposure enhanced SIAH2 expression, CSC spheroid growth, and resistance to fluorouracil, leucovorin, and oxaliplatin (FOLFOX) chemotherapy, whereas SIAH2 depletion effectively abrogated these effects. Collectively, these findings identify the SIAH2/WNK1 axis as a central metabolic regulator linking glycolysis, CSC maintenance, and microenvironment-driven therapy resistance in CRC, highlighting its potential as a therapeutic target. Full article
Show Figures

Figure 1

20 pages, 1552 KB  
Review
Engineered Mesenchymal Stromal Cells in Oncology: Navigating Between Therapeutic Delivery and Tumor Promotion
by Marta Warzycha, Agnieszka Oleksiuk, Olga Suska, Tomasz Jan Kolanowski and Natalia Rozwadowska
Genes 2026, 17(1), 108; https://doi.org/10.3390/genes17010108 - 20 Jan 2026
Abstract
Mesenchymal stromal cells (MSCs) are intensively investigated in oncology owing to their intrinsic tumor-homing ability and capacity to deliver therapeutic agents directly into the tumor microenvironment (TME). Recent advances in genetic engineering have enabled precise modification of MSCs, allowing controlled expression of therapeutic [...] Read more.
Mesenchymal stromal cells (MSCs) are intensively investigated in oncology owing to their intrinsic tumor-homing ability and capacity to deliver therapeutic agents directly into the tumor microenvironment (TME). Recent advances in genetic engineering have enabled precise modification of MSCs, allowing controlled expression of therapeutic genes and other cargo delivery, thus improving targeting efficiency. As cellular carriers, MSCs have been engineered to transport oncolytic viruses, suicide genes in gene-directed enzyme prodrug therapy (GDEPT), multifunctional nanoparticles, and therapeutic factors such as IFN-β or TRAIL, while engineered MSC-derived extracellular vesicles (MSC-EVs) offer a promising cell-free alternative. These strategies increase intratumoral drug concentration, amplify bystander effects, and synergize with standard therapies while reducing systemic toxicity. Conversely, accumulating evidence highlights the tumor-promoting properties of MSCs: once recruited by inflammatory and hypoxic cues, they remodel the tumor microenvironment by stimulating angiogenesis, suppressing immune responses, differentiating into cancer-associated fibroblasts, and promoting epithelial-to-mesenchymal transition (EMT), ultimately enhancing invasion, metastasis, and therapy resistance. This duality has sparked both enthusiasm and concern in the oncology field. The present review outlines the paradoxical role of MSCs in oncology—ranging from their potential to promote tumor growth to their emerging utility as vehicles for targeted drug delivery. By highlighting both therapeutic opportunities and biological risks, we aim to provide a balanced perspective on how MSC-based strategies might be refined, optimized, and safely integrated into future cancer therapies. Full article
(This article belongs to the Section Human Genomics and Genetic Diseases)
Show Figures

Figure 1

21 pages, 3116 KB  
Review
The Role of Cancer-Associated Fibroblasts and Tumor-Associated Macrophages in the Tumor Microenvironment and Their Impact on Ovarian Cancer Survival and Therapy
by Alena A. McQuarter, Joseph Cruz, Celina R. Yamauchi, Mariem Chouchen, Cody S. Carter, Tonya J. Webb and Salma Khan
Curr. Oncol. 2026, 33(1), 59; https://doi.org/10.3390/curroncol33010059 - 19 Jan 2026
Viewed by 10
Abstract
Ovarian cancer is the deadliest gynecologic cancer, mainly because it is often diagnosed late and resists standard treatments. The tumor microenvironment (TME) plays a major role in disease progression and therapy failure. Two key components of the TME, cancer-associated fibroblasts (CAFs) and tumor-associated [...] Read more.
Ovarian cancer is the deadliest gynecologic cancer, mainly because it is often diagnosed late and resists standard treatments. The tumor microenvironment (TME) plays a major role in disease progression and therapy failure. Two key components of the TME, cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs), create conditions that facilitate tumor growth and immune evasion. CAFs are highly diverse and originate from sources like fibroblasts and stem cells. They support cancer by remodeling the extracellular matrix, promoting angiogenesis, and releasing cytokines and growth factors that aid tumor survival. TAMs, which are usually in an M2 state, also promote metastasis and suppress immune responses by secreting immunosuppressive molecules. Together, CAFs and TAMs interact with cancer cells to activate pathways such as the TGF-β, IL-6, and PI3K/AKT pathways, which drive resistance to therapy. New treatments aim to block these interactions by targeting CAFs and TAMs through depletion, reprogramming, or pathway inhibition, often combined with immunotherapy. Advances such as single-cell sequencing and spatial transcriptomics now enable more precise identification of CAF and TAM subtypes, enabling more targeted therapies. This review summarizes their roles in epithelial ovarian cancer and explores how targeting these cells could improve outcomes. Full article
Show Figures

Figure 1

15 pages, 270 KB  
Article
Experience of a Referral Center with Desmoid Tumors, Part 2: A Retrospective Analysis of 109 Cases
by Alvarez Alvarez Rosa, Agra Pujol Carolina, Arregui Valles Marta, Alijo Francisco, Fernández Gonzalo Adriana, Gutiérrez Natalia, Lozano Lominchar Pablo, Mata Fernández Cristina, Mediavilla Santos Lydia, Novo Ulrike, Santos Marina, Hernández Torrado Guillermo, Carpintero García Henar and Gutiérrez-Ortiz de la Tabla Ana
Cancers 2026, 18(2), 305; https://doi.org/10.3390/cancers18020305 - 19 Jan 2026
Viewed by 38
Abstract
Background: Desmoid tumors (DTs) are rare, locally aggressive fibroblastic neoplasms with highly heterogeneous clinical behavior. The present work constitutes the second part of a two-part project, following our previously published multidisciplinary review of the diagnostic and therapeutic landscape of DTs. It provides a [...] Read more.
Background: Desmoid tumors (DTs) are rare, locally aggressive fibroblastic neoplasms with highly heterogeneous clinical behavior. The present work constitutes the second part of a two-part project, following our previously published multidisciplinary review of the diagnostic and therapeutic landscape of DTs. It provides a comprehensive analysis of our institutional experience as a national reference center for sarcoma. We aim to describe real-world diagnostic pathways, management strategies, and clinical outcomes in a high-volume cohort. Methods: We conducted a retrospective cohort study that included patients diagnosed with DT at our center between 2014 and 2024. Demographic, clinical, molecular, treatment, and outcome data were collected. Management strategies were analyzed according to tumor location, symptoms, progression patterns, and multidisciplinary decision-making. Outcomes included response rates, event-free survival (EFS), need for active treatment, response to systemic therapy, and recurrence after local treatments. Results: A total of 109 patients were included (median age 36.8 years; 56.9% women). Somatic CTNNB1 mutations were identified in 23 of 29 tested patients, predominantly T41A, while germline alterations were found in 18 patients, mainly in APC. Initial management was conservative in 40.4% of patients and active in 59.6%, primarily through surgery. After a median follow-up of 41.5 months, 44.9% of patients experienced disease progression. Among patients managed with active surveillance, spontaneous regression occurred in 22.2%, and 58% remained treatment-free. Surgical relapse occurred in 35.8% of patients undergoing upfront resection, with major postoperative complications limited to externally operated cases. Cryoablation achieved radiological responses in most evaluable patients, while systemic therapies showed clinical activity but relevant toxicity, particularly with tyrosine kinase inhibitors. The median EFS for the whole cohort was 57 months. Conservative initial management and R1/2 surgical margins were independently associated with worse EFS. Conclusions: Our results support a personalized, multidisciplinary management strategy for DTs, prioritizing conservative approaches when appropriate and reserving active treatments for progressive or symptomatic disease. Outcomes achieved in a specialized referral center are comparable to those reported in large international retrospective series, underscoring the value of expert multidisciplinary care in optimizing DT management. Full article
(This article belongs to the Special Issue News and How Much to Improve in Management of Soft Tissue Sarcomas)
18 pages, 8542 KB  
Article
Prehabilitation as a Biologically Active Intervention Is Associated with the Remodeling of the Pancreatic Tumor-Immune Microenvironment
by Renee Stubbins, Boris Li, Matthew Vasquez, Blythe K. Gorman, Joseph Zambelas, Kelvin Allenson, Atiya Dhala, Wenjuan Dong, Hong Zhao and Stephen Wong
Int. J. Mol. Sci. 2026, 27(2), 943; https://doi.org/10.3390/ijms27020943 - 18 Jan 2026
Viewed by 60
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is highly lethal, and many patients cannot undergo curative surgery due to frailty. Multimodal prehabilitation: combining exercise, nutrition, and psychological support improves functional readiness, but its biological impact on the PDAC tumor microenvironment (TME) is unclear. In this exploratory [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) is highly lethal, and many patients cannot undergo curative surgery due to frailty. Multimodal prehabilitation: combining exercise, nutrition, and psychological support improves functional readiness, but its biological impact on the PDAC tumor microenvironment (TME) is unclear. In this exploratory pilot study, we profiled resected PDAC tissues from prehabilitation-treated patients and matched controls using NanoString GeoMx Digital Spatial Profiling across immune, tumor, and stromal compartments (n = 4). Transcriptomic signatures were analyzed via differential expression, pathway enrichment, and MCP-counter deconvolution; protein-level validation used multiplex immunofluorescence (n = 8). Ligand–receptor modeling assessed cell–cell communication, and prognostic relevance was evaluated in TCGA-PDAC (n = 178). Prehabilitation was associated with increased NK-cell cytotoxicity, interferon response, and chemokine recruitment, as well as higher neutrophil signatures (p < 0.01) and reduced fibroblast signatures (p < 0.05). Tumor regions showed lower MAPK and PI3K/AKT activity, while stroma exhibited decreased TGF-β and Wnt signaling. Immunofluorescence confirmed neutrophil infiltration and reduced fibroblast density. TCGA analysis linked neutrophil-high/fibroblast-low profiles to longer survival (1044.6 vs. 458.7 days, p = 0.0052). These findings suggest prehabilitation may promote a more immune-active, less fibrotic TME in PDAC, resembling transcriptional states associated with improved survival. Prospective studies integrating biological and clinical endpoints are warranted. Full article
Show Figures

Graphical abstract

19 pages, 1940 KB  
Article
Protective Effect of Multifloral Honey on Stem Cell Aging in a Dynamic Cell Culture Model
by Fikriye Fulya Kavak, Sara Cruciani, Giuseppe Garroni, Diletta Serra, Rosanna Satta, Ibrahim Pirim, Melek Pehlivan and Margherita Maioli
Antioxidants 2026, 15(1), 115; https://doi.org/10.3390/antiox15010115 - 16 Jan 2026
Viewed by 282
Abstract
Natural compounds, as honey-derived flavonoids and phenolic compounds, are increasingly investigated for their potential to mitigate skin aging and prevent oxidative stress-induced cellular damages. In this context, a dynamic cell culture model was employed to assess the protective influence of honey pre-treatment on [...] Read more.
Natural compounds, as honey-derived flavonoids and phenolic compounds, are increasingly investigated for their potential to mitigate skin aging and prevent oxidative stress-induced cellular damages. In this context, a dynamic cell culture model was employed to assess the protective influence of honey pre-treatment on stem cell–associated genes and the Wingless-related integration site (Wnt) signaling pathway following ultraviolet (UV)-induced aging. Using a bioreactor, skin stem cells (SSCs) derived from healthy skin biopsies and human skin fibroblasts (HFF1) were pre-treated with 1% honey for 48 h and then exposed to UV. Real-time quantitative polymerase chain reaction (RT-qPCR) analyses were performed on Wnt signaling and anti-aging molecular responses. Honey pre-treatment enhanced the expression of pluripotency markers (Octamer-binding transcription factor 4 (Oct4); SRY-box transcription factor 2 (Sox2)) and reduced senescence-related cell cycle regulators (cyclin-dependent kinase inhibitor 2A (p16); cyclin-dependent kinase inhibitor 1A (p21); tumor protein 53 (p53)) in SSCs. In UV-damaged SSCs, honey also significantly increased Wnt3a expression. In fibroblasts, honey pre-treatment upregulated Heat shock protein 70 (Hsp70) and Hyaluronan synthase 2 (HAS2) expression, while downregulating caspase-8 (CASP8), indicating a protective role against UV-mediated cellular stress. We also analyzed nitric oxide release and the total antioxidant capacity of cells after treatment. Collectively, these findings suggest that honey may safeguard skin stem cells from UV-induced aging by modulating pluripotency and senescence-associated genes and regulating differentiation through alterations in Wnt signaling. Furthermore, Hsp70 upregulation in fibroblasts appears to strengthen cellular stress responses and support homeostatic stability. Full article
(This article belongs to the Special Issue Oxidative Stress in Cell Senescence)
Show Figures

Figure 1

22 pages, 2522 KB  
Article
Oncological Safety of High Hydrostatic Pressure Treatment: Effects on Cancer-Associated Fibroblast-like Transdifferentiation of Adipose Stromal Cells
by Julia Kristin Brach, Vivica Freiin Grote, Anika Jonitz-Heincke, Rainer Bader, Daniel Strüder, Marco Hoffmann, Sven Gerlach, Petra Fischer, Markus Wirth, Tim Ruhl, Justus P. Beier, Agmal Scherzad and Stephan Hackenberg
Curr. Issues Mol. Biol. 2026, 48(1), 91; https://doi.org/10.3390/cimb48010091 - 16 Jan 2026
Viewed by 119
Abstract
Oncological safety is essential for autologous reconstruction after resection of cartilage-infiltrating head and neck tumors. High hydrostatic pressure (HHP) enables complete devitalization of tumor-infiltrated tissue while preserving extracellular matrix integrity. However, residual soluble tumor-derived products may influence infiltrating stromal cells. This study examined [...] Read more.
Oncological safety is essential for autologous reconstruction after resection of cartilage-infiltrating head and neck tumors. High hydrostatic pressure (HHP) enables complete devitalization of tumor-infiltrated tissue while preserving extracellular matrix integrity. However, residual soluble tumor-derived products may influence infiltrating stromal cells. This study examined whether conditioned media (CM) from HHP-treated head and neck squamous cell carcinoma (HNSCC) cells induce cancer-associated fibroblast (CAF)-like transdifferentiation of human adipose stromal cells (hASCs). HASCs were exposed to CM from untreated or HHP-treated (300 MPa) HNSCC cells, tumor-CM (TCM), or TGF-β1. Morphological changes in hASCs were evaluated, and CAF marker expression was analyzed by qRT-PCR, immunofluorescence, Western blot, and ELISA. Cytokines were quantified via multiplex analysis. TGF-β1 induced a CAF-like phenotype with α-SMA upregulation, whereas TCM and 0 MPa-CM caused only modest increases in selected markers. Although 300 MPa-CM did not induce CAF-associated molecular signatures, hASCs exhibited morphological alterations, underscoring that morphology alone is insufficient to define CAF transdifferentiation. Cytokine secretion was elevated in response to all CM conditions. These findings indicate that HHP treatment at 300 MPa abolishes the paracrine CAF-inducing potential of tumor-derived mediators in vitro, supporting the oncological safety of HHP-treated tissues under these experimental condition, although further in vivo validation is warranted Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

18 pages, 14186 KB  
Article
Modulation of Cancer-Associated Fibroblasts via the miR-624-5p/FAP Axis Drives Progression and Metastasis in Non-Small Cell Lung Cancer
by Yan Zhao, Shuman Zhen, Xiaoxu Li, Xiaolin Chen, Xue Zhang, Xinming Zhao and Lihua Liu
Cancers 2026, 18(2), 279; https://doi.org/10.3390/cancers18020279 - 16 Jan 2026
Viewed by 99
Abstract
Background: Cancer-associated fibroblasts (CAFs) are key mediators of metastatic progression in non-small cell lung cancer (NSCLC). Fibroblast activation protein (FAP) serves as the hallmark of CAF activation. However, the upstream regulation of FAP remains elusive, limiting stroma-targeted therapy development. Methods: 68Ga-FAP inhibitor [...] Read more.
Background: Cancer-associated fibroblasts (CAFs) are key mediators of metastatic progression in non-small cell lung cancer (NSCLC). Fibroblast activation protein (FAP) serves as the hallmark of CAF activation. However, the upstream regulation of FAP remains elusive, limiting stroma-targeted therapy development. Methods: 68Ga-FAP inhibitor (FAPI)-04 PET/CT imaging was performed on 61 NSCLC patients to evaluate the clinical significance of FAP. CAFs and normal fibroblasts (NFs) were isolated from patient tissues. Bioinformatic analysis and qRT-PCR were employed to screen and validate miRNAs. Functional assays (CCK-8, collagen contraction, wound healing, transwell co-culture) were utilized to investigate the role of miR-624-5p in regulating fibroblast activation and the effects on the metastatic potential of NSCLC cells. The targeting relationship between miR-624-5p and FAP was validated using FISH, dual-luciferase assay, and Western blotting. Results: 68Ga-FAPI-04 uptake was higher in advanced NSCLC (p < 0.001) and correlated with tumor size, lymph node metastases, and distant metastases (p < 0.05). Isolated primary CAFs significantly enhanced the migration and invasion of A549 and PC9 cells compared to NFs (p < 0.001). We identified miR-624-5p as a significantly downregulated miRNA in CAFs (p < 0.001). Functionally, miR-624-5p overexpression inhibited CAF proliferation and collagen contraction (p < 0.01) and reduced the proliferation, migration, and invasion capabilities of A549 and PC9 cells (p < 0.001). Mechanistically, miR-624-5p bound to FAP mRNA and negatively regulated FAP expression (p < 0.001), thus suppressing CAF activation and tumor metastasis. Conclusions: Our findings establish miR-624-5p as a novel upstream regulator that suppresses FAP expression, consequently inhibiting CAF activation and its pro-metastatic function. Targeting the miR-624-5p/FAP axis represents a promising therapeutic strategy for NSCLC metastasis. Full article
(This article belongs to the Special Issue Lung Cancer: Updates on Therapy and Prognostic Prediction)
Show Figures

Figure 1

23 pages, 852 KB  
Review
Evolving Paradigms in Gastric Cancer Staging: From Conventional Imaging to Advanced MRI and Artificial Intelligence
by Giovanni Balestrucci, Vittorio Patanè, Nicoletta Giordano, Anna Russo, Fabrizio Urraro, Valerio Nardone, Salvatore Cappabianca and Alfonso Reginelli
Diagnostics 2026, 16(2), 284; https://doi.org/10.3390/diagnostics16020284 - 16 Jan 2026
Viewed by 119
Abstract
Background: Accurate preoperative staging is the cornerstone of therapeutic decision-making in gastric cancer (GC), yet standard modalities often fail to capture the full extent of disease, particularly in diffuse and poorly cohesive histotypes. This review aims to provide a comprehensive update on [...] Read more.
Background: Accurate preoperative staging is the cornerstone of therapeutic decision-making in gastric cancer (GC), yet standard modalities often fail to capture the full extent of disease, particularly in diffuse and poorly cohesive histotypes. This review aims to provide a comprehensive update on diagnostic imaging for GC, evaluating the established roles of CT, EUS, and PET/CT alongside the emerging capabilities of Magnetic Resonance Imaging (MRI) and Artificial Intelligence (AI). Methods: A structured narrative review was conducted by searching indexed biomedical databases for studies published between 2015 and 2024. A structured literature search screening process identified 410 relevant studies focusing on T, N, and M staging accuracy, quantitative imaging biomarkers, and radiomics. Results: While Multidetector CT remains the universal first-line modality, its sensitivity declines in infiltrative tumors and low-volume peritoneal carcinomatosis. EUS retains superiority for early (T1-T2) lesions but may offer limited value in advanced stages. Conversely, MRI (leveraging diffusion-weighted imaging (DWI) and multiparametric protocols) indicates superior soft-tissue contrast, potentially outperforming CT in the assessment of serosal invasion, nodal involvement, and occult peritoneal metastases. Furthermore, emerging fibroblast activation protein inhibitor (FAPI) PET tracers show promise in overcoming the limitations of FDG in mucinous and diffuse GC. Finally, radiomics and deep learning models are providing novel quantitative biomarkers for non-invasive risk stratification. Conclusions: Contemporary GC staging requires a tailored, multimodality approach. Evidence supports the increasing integration of MRI and quantitative imaging into clinical workflows to overcome the limitations of conventional techniques and support precision oncology. Full article
(This article belongs to the Special Issue Innovations in Medical Imaging for Precision Diagnostics)
Show Figures

Figure 1

25 pages, 18497 KB  
Article
Carvacrol Selectively Induces Mitochondria-Related Apoptotic Signaling in Primary Breast Cancer-Associated Fibroblasts
by Nail Besli, Nilufer Ercin, Merve Tokocin, Sümeyra Emine Boluk, Rabia Kalkan Cakmak, Kamil Ozdogan, Talar Vartanoglu Aktokmakyan, Mehtap Toprak, Gulcin Ercan, Merve Beker, Ulkan Celik, Emir Capkinoglu and Yusuf Tutar
Pharmaceuticals 2026, 19(1), 142; https://doi.org/10.3390/ph19010142 - 14 Jan 2026
Viewed by 220
Abstract
Background/Objectives: Cancer-associated fibroblasts (CAFs) are key stromal mediators of breast tumor progression and therapy resistance. Carvacrol, a dietary monoterpenic phenol, exhibits antiproliferative activity in cancer cells, but its effects on primary human breast CAFs remain unclear. This study aimed to determine whether [...] Read more.
Background/Objectives: Cancer-associated fibroblasts (CAFs) are key stromal mediators of breast tumor progression and therapy resistance. Carvacrol, a dietary monoterpenic phenol, exhibits antiproliferative activity in cancer cells, but its effects on primary human breast CAFs remain unclear. This study aimed to determine whether carvacrol selectively induces mitochondria-related apoptotic signaling in breast CAFs while sparing normal fibroblasts (NFs). Methods: Primary fibroblast cultures were established from invasive ductal carcinoma tissues (CAFs, n = 9) and nonmalignant breast tissues (NFs, n = 5) and validated by α-SMA and FAP immunofluorescence. Cells were exposed to 400 μM carvacrol. Apoptosis was assessed by TUNEL assay and BAX/BCL-XL Western blotting. Changes in signaling pathways were evaluated by analyzing PPARα/NF-κB, sirtuin (SIRT1, SIRT3), autophagy-related markers (LAMP2A, p62), and matrix metalloproteinases (MMP-2, MMP-3). In silico molecular docking and 100-ns molecular dynamics simulations were performed to examine interactions between carvacrol and caspase-3 and caspase-9. Results: Carvacrol induced a pronounced, time-dependent apoptotic response in CAFs, with TUNEL-based viability declining to approximately 10% of control levels by 12 h and a marked increase in the BAX/BCL-XL ratio. In contrast, NFs exhibited minimal TUNEL positivity and no significant change in BAX/BCL-XL. In CAFs, but not NFs, carvacrol reduced PPARα expression and NF-κB nuclear localization, increased SIRT1 and SIRT3 levels, selectively suppressed MMP-3 while partially normalizing MMP-2, and altered autophagy-related markers (decreased LAMP2A and accumulation of p62), consistent with autophagic stress and possible impairment of autophagic flux. Computational analyses revealed stable carvacrol binding to caspase-3 and caspase-9 with modest stabilization of active-site loops, supporting caspase-dependent, mitochondria-related apoptosis. Conclusions: Carvacrol selectively targets breast cancer-associated fibroblasts by inducing mitochondria-related apoptotic signaling while largely sparing normal fibroblasts. This effect is accompanied by coordinated modulation of PPARα/NF-κB, sirtuin, autophagy, and MMP pathways. These findings support further evaluation of carvacrol as a microenvironment-directed adjunct in breast cancer therapy. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Graphical abstract

33 pages, 4569 KB  
Article
Heterotypic 3D Model of Breast Cancer Based on Tumor, Stromal and Endothelial Cells: Cytokines Interaction in the Tumor Microenvironment
by Anastasia Leonteva, Alina Kazakova, Ekaterina Berezutskaya, Anna Ilyina, David Sergeevichev, Sergey Vladimirov, Maria Bogachek, Igor Vakhrushev, Pavel Makarevich, Vladimir Richter and Anna Nushtaeva
Cells 2026, 15(2), 145; https://doi.org/10.3390/cells15020145 - 14 Jan 2026
Viewed by 169
Abstract
The recreation of the tumor microenvironment remains a significant challenge in the development of experimental cancer models. The present study constitutes an investigation into the interconnection between tumor, endothelial and stromal cells in heterotypic breast cancer spheroids. The generation of models was achieved [...] Read more.
The recreation of the tumor microenvironment remains a significant challenge in the development of experimental cancer models. The present study constitutes an investigation into the interconnection between tumor, endothelial and stromal cells in heterotypic breast cancer spheroids. The generation of models was achieved through the utilization of MCF7, MDA-MB-231, and SK-BR-3 tumor cell lines, in conjunction with endothelial TIME-RFP cells and either cancer-associated (BrC4f) or normal (BN120f) fibroblasts, within ultra-low attachment plates. It was established that stromal cells, most notably fibroblasts, were conducive to the aggregation of tumor cells into spheroids and the formation of pseudovessels in close proximity to fibroblast bands. In contrast to the more aggressive tumor models MDA-MB-231 and SK-BR-3, microenvironment cells do not influence the migration ability of MCF7 tumor cells. Heterotypic spheroids incorporating CAFs demonstrated a more aggressive and immunosuppressive phenotype. Multiplex immunoassay analysis of cytokines, followed by STRING cluster analysis, was used to identify key processes including angiogenesis, invasion, stem cell maintenance, and immunosuppression. Furthermore, a cluster of cytokines (LIF, SDF-1, HGF, SCGFb) was identified as potentially involved in the regulation of PD-L1 expression by tumor cells. This finding reveals a potential mechanism of immune evasion and suggests new avenues for therapeutic investigation. Full article
(This article belongs to the Special Issue Cell-to-Cell Crosstalk as a Target of Therapies)
Show Figures

Graphical abstract

14 pages, 5733 KB  
Article
Immunohistochemical Pattern of CD34 Distribution in Different Types of Basal Cell Carcinoma and in Peritumoral Skin
by Vladimir Petrovic, Aleksandar Petrovic, Ivan R. Nikolic, Nataša Vidovic, Tijana Dencic, Ilija Golubovic, Miroslav Milic and Aleksandra Antovic
Medicina 2026, 62(1), 158; https://doi.org/10.3390/medicina62010158 - 13 Jan 2026
Viewed by 165
Abstract
Background and Objectives: Basal cell carcinoma (BCC) is the most common skin carcinoma, mainly occurring in older individuals. The aim of this study was to document the immunohistochemical distribution of CD34 in different histopathological types of BCC, as well as in the [...] Read more.
Background and Objectives: Basal cell carcinoma (BCC) is the most common skin carcinoma, mainly occurring in older individuals. The aim of this study was to document the immunohistochemical distribution of CD34 in different histopathological types of BCC, as well as in the peritumoral and uninvolved skin of biopsy samples. Materials and Methods: Excisional biopsies of skin BCCs were routinely processed into paraffin blocks, and microtome sections were stained immunohistochemically for CD34. Results: A consistent finding in skin samples containing BCC was the absence of CD34 in the following extravascular structures: neoplastic cells, epidermis and its derivatives (except for the cells of the isthmic part of the outer hair follicle sheath), fibroblast-like cells of BCC tumor stroma, as well as in the papillary dermis in the tumor region. Fibroblast-like cells of the tumor stroma were variably CD34 immunopositive only in the nodular type of BCC. In all examined biopsies, part of the dermis adjacent to the BCC tumor mass (juxtatumoral zone) was characterized by pronounced CD34 immunopositivity. In the transitional zone of peritumoral skin and in marginal skin, CD34-positive connective tissue cells were observed in the periadnexal dermis around: sebaceous gland lobules, the secretory coils of eccrine sweat glands, the pilosebaceous canal, as well as in the perimysium of the arrector pili muscle. Fibrocytes of fibrous sheaths encasing the isthmic part of hair follicles were CD34 negative, interposed between highly positive epithelial cells of the outer hair follicle sheath and the fibroblasts of the local reticular dermis. The transitional zone and uninvolved skin contained CD34-positive fibroblast-like cells situated between secondary bundles of reticular dermis, as well as CD34-positive cell processes within these bundles. Conclusions: The observed pattern of CD34 positivity within the examined regions shows a specific distribution, providing insight into the adaptive responses of the skin to the tumoral process. Full article
(This article belongs to the Section Oncology)
Show Figures

Figure 1

21 pages, 78949 KB  
Article
FGF2 as a Potential Tumor Suppressor in Lung Adenocarcinoma
by Shih-Sen Lin, Hsin-Ying Lu, Tsung-Ming Chang, Ying-Sui Sun and Ju-Fang Liu
Diagnostics 2026, 16(2), 250; https://doi.org/10.3390/diagnostics16020250 - 13 Jan 2026
Viewed by 235
Abstract
Background/Objectives: Lung adenocarcinoma (LUAD), the predominant subtype of non-small cell lung cancer (NSCLC), is frequently diagnosed at advanced stages with distant metastasis, underscoring the need for effective prognostic biomarkers. Fibroblast growth factor 2 (FGF2), a multifunctional regulator, has shown to play contradictory [...] Read more.
Background/Objectives: Lung adenocarcinoma (LUAD), the predominant subtype of non-small cell lung cancer (NSCLC), is frequently diagnosed at advanced stages with distant metastasis, underscoring the need for effective prognostic biomarkers. Fibroblast growth factor 2 (FGF2), a multifunctional regulator, has shown to play contradictory roles in cancer progression. Methods: We analyzed three independent Gene Expression Omnibus (GEO) datasets (GSE19804, GSE18842, and GSE19188) to identify consistently dysregulated genes in LUAD. Functional enrichment (GO, KEGG, and cancer hallmark analysis), protein–protein interaction (PPI) network construction, and hub gene prioritization were performed using public bioinformatic tools. Survival analyses were conducted via the Kaplan–Meier Plotter. The expression of FGF2 was validated across multiple platforms, including TCGA, CPTAC, TNMplot, LCE, and the Human Protein Atlas. Functional assays (Transwell migration and wound healing) demonstrated that exogenous FGF2 significantly suppressed LUAD cell motility in vitro. Results: A total of 949 differentially expressed genes (DEGs) were commonly identified across datasets, with enrichment in cell adhesion and metastasis-related pathways. Among the 11 hub genes identified, FGF2 was consistently downregulated in LUAD tissues across all datasets and stages. Higher FGF2 expression was associated with longer overall and progression-free survival. In vitro, FGF2 treatment significantly suppressed the migration and wound healing abilities of LUAD cell lines. Conclusions: FGF2 is downregulated in LUAD and inversely associated with metastatic progression and poor prognosis. The observed reduction in cancer cell motility upon FGF2 treatment in vitro, together with its expression pattern, supports a potential tumor-suppressive role and suggests that FGF2 may serve as a candidate non-invasive biomarker for monitoring LUAD metastasis. Full article
Show Figures

Figure 1

19 pages, 1296 KB  
Article
68Ga-FAPI-04 PET/CT in the Diagnosis of Hepatocellular Carcinoma Associated with Cirrhosis: Diagnostic Value, Correlation Between PET Parameters of the Tumor and Its Size, and PIVKA-II Levels
by Zhamilya Zholdybay, Zhanar Zhakenova, Bekzhan Issamatov, Madina Gabdullina, Yevgeniya Filippenko, Suriya Yessentayeva, Galymzhan Alisherov, Jandos Amankulov and Ildar Fakhradiyev
Diagnostics 2026, 16(2), 249; https://doi.org/10.3390/diagnostics16020249 - 13 Jan 2026
Viewed by 224
Abstract
Background/Objectives: Hepatocellular carcinoma remains a major cause of death from cancer globally. While 18F-FDG PET/CT is commonly used for tumor imaging, its sensitivity is limited, especially due to high liver background uptake. Recently, 68Ga-FAPI PET/CT, which targets fibroblast activation protein in [...] Read more.
Background/Objectives: Hepatocellular carcinoma remains a major cause of death from cancer globally. While 18F-FDG PET/CT is commonly used for tumor imaging, its sensitivity is limited, especially due to high liver background uptake. Recently, 68Ga-FAPI PET/CT, which targets fibroblast activation protein in tumor stroma, has emerged as a promising diagnostic tool. In this study, we aimed to assess the diagnostic performance of 68Ga-FAPI-04 PET/CT in HCC patients with and without liver cirrhosis and to explore the relationship between PET metrics, tumor size, and PIVKA-II serum marker. Methods: In this prospective single-center study, 59 patients with confirmed HCC (37 with cirrhosis, 22 without) underwent 68Ga-FAPI-04 PET/CT. The standard dose (1.5–2.0 MBq/kg) was administered intravenously, and imaging was carried out 60 min post-injection. Semi-quantitative parameters including SUVmax, SUVmean, and tumor-to-background ratio were calculated. Diagnostic performance was assessed using histopathology and multimodal imaging. Statistical analyses included the Mann–Whitney U test and Spearman correlation. Results: The overall sensitivity for HCC detection was 89.8%, with a specificity of 60% and accuracy of 87%. Sensitivity and specificity showed a tendency to be lower in cirrhotic compared with non-cirrhotic patients, with a notably higher background liver uptake in cirrhosis (SUVmax 3.60 vs. 1.3, p < 0.001), resulting in lower TBR values (3.7 vs. 7.0, p < 0.001). A strong correlation between SUVmax and tumor size was seen in non-cirrhotic HCC, while a moderate association between SUVmax and PIVKA-II levels was observed in cirrhotic patients. Conclusions:68Ga-FAPI-04 PET/CT demonstrates high sensitivity for HCC detection and may serve as a complementary imaging modality, particularly when interpreted through conventional cross-sectional imaging. Image interpretation in cirrhotic livers may be challenging due to increased background uptake and reduced TBR. Associations between PET-derived parameters, tumor size, and serum PIVKA-II levels should be considered hypothesis-generating and require validation in larger, multicenter studies with clinical outcome data. Full article
(This article belongs to the Collection Nuclear Medicine and Molecular Imaging Technology)
Show Figures

Figure 1

21 pages, 13737 KB  
Article
Adipose Stromal Cell-Derived Cancer-Associated Fibroblasts Promote Pancreatic Adenocarcinoma Progression Through SFRP4 Signaling
by Joseph Rupert, Lingyi Cai, Alexes C. Daquinag, Dimitris Anastassiou and Mikhail G. Kolonin
Cancers 2026, 18(2), 233; https://doi.org/10.3390/cancers18020233 - 12 Jan 2026
Viewed by 236
Abstract
Background/objectives: Progression of pancreatic ductal adenocarcinoma (PDAC) and other carcinomas relies on cancer-associated fibroblasts (CAFs). A subset of CAFs is derived from adipose stromal cells (ASCs) recruited by tumors and the ASC-CAF conversion has been associated with invasiveness and poor prognosis. Methods: To [...] Read more.
Background/objectives: Progression of pancreatic ductal adenocarcinoma (PDAC) and other carcinomas relies on cancer-associated fibroblasts (CAFs). A subset of CAFs is derived from adipose stromal cells (ASCs) recruited by tumors and the ASC-CAF conversion has been associated with invasiveness and poor prognosis. Methods: To explore the underlying molecular mechanisms, we used a model based on primary ASCs derived from human visceral adipose tissue co-cultured with human PDAC cell line Capan-1. To investigate cancer progression in vivo, we also used mice orthotopically grafted with mouse KPC cells. Results: Genomic analysis revealed that Capan-1 co-culture induces Wnt and TGFβ signaling and extracellular matrix (ECM) gene expression in ASC. We investigated the function of two markers of the fibroblastic transition highly induced by cancer cells: a long non-coding RNA LINC01614 and a Wnt signaling modulator SFRP4. By using ASCs with either SFRP4 or LINC01614 knocked out (ko), we showed that both genes are required for Wnt/TGFβ signaling and ECM induction in ASCs by Capan-1. Analysis of changes in Capan-1 genes that rely on LINC01614 and SFRP4 expression in ASCs also identified the Wnt and TGF pathways. SFRP4 ko in ASCs suppressed both migration and invasion of Capan-1 cells. We show that tumors in SFRP4 ko mice have less desmoplasia, less epithelial dedifferentiation, reduced growth rate, and reduced progression to metastasis. Conclusions: We conclude that SFRP4 promotes cancer progression in pancreatic cancer and is a promising therapeutic target. Full article
Show Figures

Figure 1

Back to TopTop