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
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (503)

Search Parameters:
Keywords = 3D tumor spheroids

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
38 pages, 839 KB  
Review
Ex Vivo Treatment Response Prediction in Multiple Myeloma: Assay Formats, Clinical Correlation, and Future Directions
by Gavin R. Oliver, Carlton C. Barnett, Kendra E. Hightower, Yubin Kang and Muhamed Baljevic
Cancers 2026, 18(3), 411; https://doi.org/10.3390/cancers18030411 - 28 Jan 2026
Viewed by 40
Abstract
Ex vivo functional testing for multiple myeloma is rapidly evolving, yet no single assay has reached the level of reliability and clinical utility needed for routine decision-making. Existing approaches generally fall into three categories: 2D cultures, 3D models, and dynamic systems. Each contributes [...] Read more.
Ex vivo functional testing for multiple myeloma is rapidly evolving, yet no single assay has reached the level of reliability and clinical utility needed for routine decision-making. Existing approaches generally fall into three categories: 2D cultures, 3D models, and dynamic systems. Each contributes valuable but incomplete insight into therapeutic response. Among these, 2D assays remain the most mature, with the most extensive clinical correlations to date, though their simplified architecture limits their ability to reflect the full complexity of the bone marrow microenvironment. However, 3D systems, including spheroids and matrix-based organoids, offer improved preservation of tumor heterogeneity and microenvironmental cues. These platforms show emerging clinical relevance and may hold advantages over traditional 2D formats, and validation efforts are developing. Dynamic systems, including microfluidic models and perfused bone-marrow mimetics, represent the most physiologically ambitious category, yet their technical intricacy and early stage of development have so far limited broad clinical correlation. Altogether, the current landscape highlights substantial progress but lacks an optimal assay. In this review, we take the unique approach of examining published ex vivo tests that have demonstrated a level of clinical correlation. We evaluate their respective formats, strengths and limitations, and discuss considerations for what an ideal future assay may encompass. Full article
(This article belongs to the Special Issue Clinical Trials and Translational Research in Multiple Myeloma)
Show Figures

Figure 1

19 pages, 10734 KB  
Article
Three-Dimensional Tumor Spheroids Reveal B7-H3 CAR T Cell Infiltration Dynamics and Microenvironment-Induced Functional Reprogramming in Solid Tumors
by Feng Chen, Ke Ning, Yuanyuan Xie, Xiaoyan Yang, Ling Yu and Xinhui Wang
Cells 2026, 15(2), 169; https://doi.org/10.3390/cells15020169 - 16 Jan 2026
Viewed by 202
Abstract
Chimeric antigen receptor (CAR) T cell therapy has demonstrated clinical success in hematologic malignancies but has limited efficacy in solid tumors due to tumor microenvironment (TME) barriers that impede CAR T cell recognition, infiltration, and sustained function. Traditional 2D assays inadequately recapitulate these [...] Read more.
Chimeric antigen receptor (CAR) T cell therapy has demonstrated clinical success in hematologic malignancies but has limited efficacy in solid tumors due to tumor microenvironment (TME) barriers that impede CAR T cell recognition, infiltration, and sustained function. Traditional 2D assays inadequately recapitulate these constraints, necessitating improved in vitro models. This study validated a 3D tumor spheroid platform using an agarose microwell system to generate uniform B7-H3-positive spheroids from multiple solid tumor cell lines, enabling the evaluation of CAR T cell activity. TME-relevant immune modulation under 3D conditions was analyzed by flow cytometry for B7-H3, MHC I/II, and antigen processing machinery (APM), followed by co-culture with B7-H3 CAR T cells to assess cytotoxicity, spheroid integrity, tumor viability, and CAR T cell activation, exhaustion, and cytokine production. Two human cancer-cell-line-derived spheroids, DU 145 (prostate cancer) and SUM159 (breast cancer), retained B7-H3 expression, while MC38 (mouse colon cancer)-derived spheroids served as a B7-H3 negative control. Under 3D culture conditions, DU 145 and SUM159 spheroids acquire TME-like immune evasion characteristics and specifically downregulated MHC-I and APM (TAP1, TAP2, LMP7) with concurrent upregulation of MHC-II and calreticulin. Co-culture showed effective spheroid infiltration, cytotoxicity, and structural disruption, with infiltrating CAR T cells displaying higher CD4+ fraction, activation, exhaustion, effector/terminal differentiation, and IFN-γ/TNF-α production. This 3D platform recapitulates critical TME constraints and provides a cost-effective, feasible preclinical tool to assess CAR T therapies beyond conventional 2D assays. Full article
(This article belongs to the Section Cell Methods)
Show Figures

Figure 1

24 pages, 3083 KB  
Article
Altered Magnesium Environments Restrict Colorectal HT-29 Spheroid Growth by Disturbing Cellular Mg2+ Homeostasis
by Nattida Kampuang, Pongsakorn Lapchock, Tanida Treerattanakulporn, Phossawee Kongkaew, Siriporn Chamniansawat and Narongrit Thongon
Int. J. Mol. Sci. 2026, 27(2), 834; https://doi.org/10.3390/ijms27020834 - 14 Jan 2026
Viewed by 136
Abstract
Dysregulated magnesium (Mg2+) homeostasis contributes to colorectal cancer (CRC), yet its context-dependent function within the tumor microenvironment remains unresolved. This study aimed to determine how sustained low and high extracellular Mg2+ environments affect CRC spheroid (SP) growth and Mg2+ [...] Read more.
Dysregulated magnesium (Mg2+) homeostasis contributes to colorectal cancer (CRC), yet its context-dependent function within the tumor microenvironment remains unresolved. This study aimed to determine how sustained low and high extracellular Mg2+ environments affect CRC spheroid (SP) growth and Mg2+ homeostasis using HT-29 SPs. We analyzed Mg2+ flux, the expression of Mg2+ transporters (e.g., Transient Receptor Potential Melastatin (TRPM) 6), viability, apoptotic and autophagic markers, and phospho-/oxidoproteomic alterations. Both Mg2+ extremes destabilized SP architecture, reduced viability, and induced apoptosis and autophagy, with SPs displaying heightened vulnerability relative to 2D cultures. Mg2+ stress impaired Mg2+ influx and eliminated adaptive transporter regulation in SPs. Loss of membrane TRPM6/7 heterodimers, driven by altered phosphorylation (e.g., TRPM6 Serine 141, Serine 1252, Threonine 1851) and elevated oxidation (e.g., Methionine 1755), suppressed channel activity. High Mg2+ caused profound metabolic failure despite increased total Mg2+, reflecting functional Mg2+ deficiency. CRC spheroids are acutely susceptible to Mg2+ imbalance due to collapsed transporter homeostasis and post-translational inhibition of Mg2+ channels. These findings reveal a targetable metabolic vulnerability and support the therapeutic potential of localized Mg2+ modulation in CRC. Full article
(This article belongs to the Special Issue The Role of Mg Homeostasis in Disease: 2nd Edition)
Show Figures

Figure 1

17 pages, 8188 KB  
Article
Leptin Drives Breast Cancer Aggressiveness Acting Through the Activation of the NCOA1/STAT3 Pathway
by Khouloud Ayed, Amal Gorrab, Hichem Bouguerra, Rym Akrout, Sami Zekri, Wassim Y. Almawi, Rahma Boughriba, Khalil Choukri, Dhouha Bacha, Alessandra Pagano, Jean-François Louet, Hervé Kovacic, Mounia Tannour-Louet and Asma Gati
Med. Sci. 2026, 14(1), 32; https://doi.org/10.3390/medsci14010032 - 8 Jan 2026
Viewed by 351
Abstract
Background/Objectives: Obesity-associated hyperleptinemia has been linked to breast cancer (BC) progression via mechanisms that remain incompletely understood. This study explores the role of leptin and its receptor (LEPR) in facilitating BC cell proliferation, migration, epithelial–mesenchymal transition (EMT), and STAT3 signaling pathway activation. [...] Read more.
Background/Objectives: Obesity-associated hyperleptinemia has been linked to breast cancer (BC) progression via mechanisms that remain incompletely understood. This study explores the role of leptin and its receptor (LEPR) in facilitating BC cell proliferation, migration, epithelial–mesenchymal transition (EMT), and STAT3 signaling pathway activation. Methods: We analyzed gene expression and survival data from TCGA BRCA dataset. MCF-7 and MDA-MB-231 BC cells were exposed to leptin at 10 ng/mL (lean-associated levels) and 100 ng/mL (elevated levels linked to obesity). MTT assays, colony formation tests, wound-healing and tumor spheroid dissemination experiments evaluated cell proliferation and migration. Immunofluorescence and Western blot analysis assessed changes in EMT markers and cytoskeletal alterations, while Western blotting and qPCR assessed STAT3 and NCOA1 expression and activation levels. Results: Elevated LEPR expression was linked with unfavorable prognosis in BC patients. Higher doses of leptin (100 ng/mL) significantly enhanced cellular proliferation rates and migratory capabilities, in both cell lines, and promoted EMT characteristics marked by downregulated E-cadherin and cytoskeleton structural changes. Whereas heightened JAK2/STAT3 signaling correlated with elevated leptin dosages, STAT3 inhibition using AG490 reversed leptin-induced migration while reinstating E-cadherin levels to baseline. Furthermore, leptin upregulated NCOA1, an essential STAT3 coactivator, facilitating increased expression of Cyclin D1 and VEGF target genes. Clinical positive relationships were seen between LEP/LEPR expressions and NCOA1 levels and between NCOA1 and various gene signatures related to STAT3/P-STAT3 within BC specimens. Conclusions: Obesity-associated hyperleptinemia enhances aggressiveness in BC through a mechanism involving LEPR-mediated activation pathways encompassing NCOA1/STAT3, which drive proliferation, migration, and EMT. This assigns a potential therapeutic utility for obesity-related advancements found within BC pathology. Full article
(This article belongs to the Special Issue Feature Papers in Section “Cancer and Cancer-Related Research”)
Show Figures

Graphical abstract

16 pages, 3831 KB  
Article
Development of a Microwell System for Reproducible Formation of Homogeneous Cell Spheroids
by Miguel A. Reina Mahecha, Ginevra Mariani, Pauline E. M. van Schaik, Paulien Schaafsma, Theo G. van Kooten, Prashant K. Sharma and Inge S. Zuhorn
Pharmaceutics 2026, 18(1), 56; https://doi.org/10.3390/pharmaceutics18010056 - 31 Dec 2025
Viewed by 415
Abstract
Background/Objectives: Three-dimensional (3D) cell cultures are increasingly used because 3D cell aggregates better mimic tissue-level biological mechanisms and support studies of tissue physiology and drug screening. However, existing laboratory methods and commercial microwell platforms often yield inconsistent results and can be error-prone, time-consuming, [...] Read more.
Background/Objectives: Three-dimensional (3D) cell cultures are increasingly used because 3D cell aggregates better mimic tissue-level biological mechanisms and support studies of tissue physiology and drug screening. However, existing laboratory methods and commercial microwell platforms often yield inconsistent results and can be error-prone, time-consuming, or costly. The objective of this work was to develop a reproducible, high-yield, and cost-effective approach for generating homogeneous cell aggregates using custom 3D-printed microwell stamps. Methods: Custom conical and semi-spherical microwell stamps were fabricated using 3D printing. Stamp resolution was characterized by scanning electron microscopy (SEM). Negative imprints were cast in polydimethylsiloxane (PDMS), a biocompatible and hydrophobic polymer conducive to cell aggregation. These PDMS microwells were then used to generate pluripotent stem cell aggregates (embryoid bodies, EBs) and tumor spheroids from adherent cancer cell lines. Results: The 3D-printed stamps produced high-resolution conical and semi-spherical microwells in PDMS. Semi-spherical microwells enabled rapid, simple, and cost-effective formation of pluripotent stem cell aggregates that were homogeneous in size and shape. These aggregates outperformed those produced using commercial microwell plates and ultra-low attachment plates. The fabricated microwells also generated uniform tumor spheroids from adherent cancer cells, demonstrating their versatility. Conclusions: The in-house 3D-printed microwell stamps offer a reproducible, efficient, and economical platform for producing homogeneous cell aggregates. This system improves upon commercial alternatives and supports a broad range of applications, including pluripotent stem cell embryoid body formation and tumor spheroid generation. Full article
(This article belongs to the Special Issue 3D Printing in Bioengineering and Pharmaceutical Manufacturing)
Show Figures

Graphical abstract

36 pages, 6557 KB  
Review
New Knowledge About Tissue Engineering Under Microgravity Conditions in Space and on Earth
by Markus Wehland, Thomas J. Corydon, Luis Fernando González-Torres, Fatima Abdelfattah, Jayashree Sahana, Herbert Schulz, Ashwini Mushunuri, Hanna Burenkova, Simon L. Wuest, Marcus Krüger, Armin Kraus and Daniela Grimm
Int. J. Mol. Sci. 2026, 27(1), 341; https://doi.org/10.3390/ijms27010341 - 28 Dec 2025
Viewed by 774
Abstract
Microgravity (µg)-generated three-dimensional (3D) multicellular aggregates can serve as models of tissue and disease development. They are relevant in the fields of cancer and in vitro metastasis or regenerative medicine (tissue engineering). Driven by the 3R concept—replacement, reduction, and refinement of [...] Read more.
Microgravity (µg)-generated three-dimensional (3D) multicellular aggregates can serve as models of tissue and disease development. They are relevant in the fields of cancer and in vitro metastasis or regenerative medicine (tissue engineering). Driven by the 3R concept—replacement, reduction, and refinement of animal testing—µg-exposure of human cells represents a new alternative method that avoids animal experiments entirely. New Approach Methodologies (NAMs) are used in biomedical research, pharmacology, toxicology, cancer research, radiotherapy, and translational regenerative medicine. Various types of human cells grow as 3D spheroids or organoids when exposed to µg-conditions provided by µg simulating instruments on Earth. Examples for such µg-simulators are the Rotating Wall Vessel, the Random Positioning Machine, and the 2D or 3D clinostat. This review summarizes the most recent literature focusing on µg-engineered tissues. We are discussing all reports examining different tumor cell types from breast, lung, thyroid, prostate, and gastrointestinal cancers. Moreover, we are focusing on µg-generated spheroids and organoids derived from healthy cells like chondrocytes, stem cells, bone cells, endothelial cells, and cardiovascular cells. The obtained data from NAMs and µg-experiments clearly imply that they can support translational medicine on Earth. Full article
Show Figures

Figure 1

25 pages, 5307 KB  
Article
Fibrillarin Contributes to the Oncogenic Characteristics of Colorectal Cancer Cells and Reduces Sensitivity to 5-Fluorouracil
by Ting Wu, Mounira Chalabi-Dchar, Wei Xiong, Lucie Arnould, Eliezer Aimontche, Sabine Beaumel, Charles Dumontet, Virginie Marcel, Tanguy Fenouil, Jean-Jacques Diaz, Marie Alexandra Albaret and Hichem Claude Mertani
Cancers 2025, 17(24), 3900; https://doi.org/10.3390/cancers17243900 - 5 Dec 2025
Viewed by 424
Abstract
Background/Objectives: Fibrillarin (FBL) is a key nucleolar methyltransferase involved in ribosome biogenesis through 2′-O-ribose methylation of rRNA. While its oncogenic role has been reported in several cancer types, its expression and function in human colorectal cancer (CRC) have remained largely unexplored. This study [...] Read more.
Background/Objectives: Fibrillarin (FBL) is a key nucleolar methyltransferase involved in ribosome biogenesis through 2′-O-ribose methylation of rRNA. While its oncogenic role has been reported in several cancer types, its expression and function in human colorectal cancer (CRC) have remained largely unexplored. This study aims to investigate the expression of FBL in human CRC tissues and cell lines and to determine its functional role in tumor progression and metastasis. Methods: We examined FBL expression in paired human CRC primary tumors and liver metastases using immunohistochemistry. Functional studies were performed using SW-480 (primary tumor) and SW-620 (lymph node metastasis) CRC cell lines derived from the same patient. Cell migration, invasion, and 3D spheroid growth were analyzed following FBL downregulation. In vivo tumor growth was assessed in SCID mice xenografted with FBL-deficient cells. Molecular changes were explored through phosphorylation arrays and Western blotting. Results: FBL expression was significantly higher in human metastatic lesions than in primary tumors. FBL downregulation impaired migration, invasion, and spheroid growth in SW-480 and SW-620 cells and reduced tumor growth in vivo. Mechanistically, FBL inhibition decreased activation of MAPK/ERK, PI3K/AKT, and JNK/p38 pathways and reduced phosphorylation of the transcription factor CREB. Conclusions: Our study identifies FBL as a potential contributor to colorectal cancer progression, with elevated expression associated particularly with metastatic disease. By demonstrating that FBL expression is elevated in patient-derived metastatic tissues and functionally promotes migration, invasion, and tumor growth, our findings expand the role of ribosome biogenesis factors beyond protein synthesis. The observed suppression of key oncogenic pathways and CREB phosphorylation upon FBL inhibition suggests that FBL integrates ribosomal regulation with cancer cell signaling. These insights open new avenues for targeting nucleolar activity in advanced CRC and highlight FBL as a potential biomarker and therapeutic target in metastatic disease. Full article
(This article belongs to the Special Issue Colorectal Cancer Liver Metastases)
Show Figures

Figure 1

10 pages, 3316 KB  
Case Report
Role of 68Ga-DOTATOC Positron Emission Tomography in Locating Pulmonary Neuroendocrine Tumor Presenting with ACTH-Dependent Cushing’s Syndrome: A Case Report
by Misako Tanaka, Masakazu Uejima, Kuniaki Ozaki, Maiko Nishigori, Yukako Kurematsu, Kosuke Kaji, Kei Moriya, Tadashi Namisaki, Akira Mitoro, Fumihiko Nishimura, Motoaki Yasukawa and Hitoshi Yoshiji
J. Clin. Med. 2025, 14(24), 8634; https://doi.org/10.3390/jcm14248634 - 5 Dec 2025
Viewed by 379
Abstract
Background: In ectopic adrenocorticotropic hormone (ACTH) syndrome, locating the responsible lesion is often challenging. Case Presentation: A 68-year-old woman was transferred to Nara Medical University hospital for a detailed investigation of her ACTH-dependent Cushing’s syndrome. Because of hypercortisolism-induced immunosuppression, she subsequently developed [...] Read more.
Background: In ectopic adrenocorticotropic hormone (ACTH) syndrome, locating the responsible lesion is often challenging. Case Presentation: A 68-year-old woman was transferred to Nara Medical University hospital for a detailed investigation of her ACTH-dependent Cushing’s syndrome. Because of hypercortisolism-induced immunosuppression, she subsequently developed severe Nocardia pneumonia and was forced to temporarily depend on noninvasive positive pressure ventilation (NIPPV). Intravenous antifungal agents and antibiotics were administered, resulting in significant symptomatic improvement. Metyrapone was administered to suppress excessive cortisol. Contrast-enhanced magnetic resonance imaging of the pituitary revealed a 4 mm sized poorly enhanced area, and microadenoma was suspected. Although cavernous venous sampling was indispensable prior to trans-spheroidal surgery (TSS), this examination could not be performed because of the presence of deep vein thrombosis. TSS was performed for both diagnostic and therapeutic purposes, but hypercortisolism did not improve. Moreover, immunohistochemical findings of the specimen revealed nonfunctional pituitary tumor. Methods: We re-evaluated the responsible lesion causing ACTH-dependent Cushing’s syndrome. Fluorine-18 fluorodeoxyglucose (FDG) positron emission tomography (PET) revealed weak and abnormal FDG uptake in the right pericardium, but the possibility of nonspecific uptake could not be ruled out. However, gallium-68 1,4,7,10-tetraazacyclododecane-N,N′,N′′,N′′′-tetraacetic-acid-D-Phe1-Tyr3-octreotide (68Ga-DOTATOC)-PET demonstrated the same degree of abnormal uptake; therefore, a functional pulmonary tumor was strongly suspected. Results: Video-Assisted Thoracic Surgery (VATS) was performed, and histopathological findings of the specimen revealed a neuroendocrine tumor with positive ACTH staining. After VATS, ACTH and cortisol levels were normalized. Conclusions: Here, we report a case of ACTH-dependent Cushing’s syndrome caused by a lung neuroendocrine tumor, in which 68Ga-DOTATOC PET was helpful in detecting the functional tumors. Full article
(This article belongs to the Section Endocrinology & Metabolism)
Show Figures

Figure 1

20 pages, 3515 KB  
Article
SOX2/SOX17 Molecular Switching by Polyphenols to Promote Thyroid Differentiation in 2D and 3D Models of Anaplastic Thyroid Cancer
by Fabiola Vaglica, Mattia Biondo, Giuseppe Siragusa, Giorgio Arnaldi, Valentina Guarnotta, Giuseppe Pizzolanti and Laura Tomasello
Biology 2025, 14(12), 1730; https://doi.org/10.3390/biology14121730 - 2 Dec 2025
Viewed by 519
Abstract
Deep alterations in tumor cell gene profiles resulting in the loss of their specific functions are frequently the cause of resistance to traditional cancer treatments. Therefore, reprogramming the expression pattern of cancer cells toward a differentiated phenotype represents a promising therapeutic strategy. In [...] Read more.
Deep alterations in tumor cell gene profiles resulting in the loss of their specific functions are frequently the cause of resistance to traditional cancer treatments. Therefore, reprogramming the expression pattern of cancer cells toward a differentiated phenotype represents a promising therapeutic strategy. In this study, we investigated whether resveratrol (RSV) and its natural analogs—3,4′,5-trimethoxystilbene (3-MET-OX) and isorhapontigenin (ISOR-H-PG)—can modulate the SOX2/SOX17 balance and promote re-differentiation in anaplastic thyroid cancer (ATC) cells. Two human ATC cell lines (SW1736 and 8505c) and non-tumoral thyroid cells (Nthy-ori 3-1) were cultured in two-dimensional (2D) or three-dimensional (3D) systems and treated with polyphenols at sub-cytotoxic doses. In 2D cultures, cell viability and cell cycle analyses confirmed a cytostatic effect characterized by G1 arrest. In 3D cultures, polyphenol treatment caused morphological disruption of ATC spheroids and significantly modulated the gene expression profile. RSV and 3-MET-OX reduced stemness markers (SOX2, NANOG), increased the thyroid lineage transcription factor (SOX17), and enhanced differentiation genes (TTF-1, TPO, NIS). Overall, these results support our hypothesis that modulation of the SOX2/SOX17 ratio by polyphenols provides a mechanistic basis for re-differentiation, thereby improving therapeutic responsiveness in ATC. Full article
Show Figures

Figure 1

22 pages, 8429 KB  
Article
SOX18 and SOX30 in NSCLC: The Epigenetic Landscape of Methylation, miRNA Regulation, and Network Crosstalk in Tumor Progression
by Mateusz Olbromski, Aleksandra Piotrowska, Monika Mrozowska, Alicja Kmiecik, Natalia Glatzel-Plucinska, Agnieszka Gomulkiewicz, Aleksandra Stepien, Klaudia Krawczynska, Piotr Blasiak, Marzenna Podhorska-Okolow and Piotr Dziegiel
Int. J. Mol. Sci. 2025, 26(23), 11669; https://doi.org/10.3390/ijms262311669 - 2 Dec 2025
Viewed by 1956
Abstract
SOX (SRY-related HMG-box) transcription factors are key regulators of embryogenesis and vascular development, with emerging roles in cancer biology. In non-small-cell lung cancer (NSCLC), the contributions of SOX18 and SOX30 remain insufficiently understood, particularly regarding their epigenetic regulation and network interactions with angiogenic [...] Read more.
SOX (SRY-related HMG-box) transcription factors are key regulators of embryogenesis and vascular development, with emerging roles in cancer biology. In non-small-cell lung cancer (NSCLC), the contributions of SOX18 and SOX30 remain insufficiently understood, particularly regarding their epigenetic regulation and network interactions with angiogenic and immune-modulatory pathways. We examined 800 NSCLC specimens (400 lung adenocarcinomas, 400 squamous cell carcinomas) using immunohistochemistry, RT-qPCR, Western blotting, and spatial transcriptomics to profile SOX18, SOX30, and related signaling partners (SOX7, SOX17, MEF2C—Myocyte Enhancer Factor 2C, VCAM1—Vascular Cell Adhesion Molecule 1, p-STAT3—Signal Transducer and Activator of Transcription 3). Epigenetic regulation was assessed via droplet digital methylation-specific PCR of promoter CpG islands, while functional validation employed adenoviral delivery of hsa-miR-24-3p in NSCLC cell lines and 3D spheroid cultures. SOX18 protein was markedly overexpressed in both NSCLC subtypes, despite reduced transcript levels and consistent promoter hypermethylation, suggesting post-transcriptional regulation. In contrast, SOX30 expression was uniformly downregulated at both mRNA and protein levels, frequently linked to promoter hypermethylation, especially in squamous carcinoma. Spatial transcriptomics revealed SOX18 enrichment at tumor cores and invasive borders, co-localizing with MEF2C, VCAM1, and p-STAT3 in vascular and stromal niches, while SOX30 expression remained low across all tumor regions. Functional assays demonstrated that hsa-miR-24-3p suppressed SOX18 expression and partially modulated SOX30 and MEF2C, reinforcing a miRNA-driven regulatory axis. In summary, SOX18 and SOX30 play divergent roles in NSCLC progression: SOX18 functions as a pro-oncogenic factor driving angiogenesis and tumor–stroma interactions, while SOX30 acts as an epigenetically silenced tumor suppressor. Regulation of SOX18 by miR-24-3p highlights a potential therapeutic vulnerability. These findings underscore the significance of SOX transcription factors as biomarkers and potential targets for novel treatment strategies in NSCLC. Full article
(This article belongs to the Special Issue Advancements in Cancer Biomarkers)
Show Figures

Graphical abstract

21 pages, 7221 KB  
Article
ADGRG6 Promotes Pancreatic Adenocarcinoma Progression Through the NF-κB/STAT6 Axis and Modulation of the Tumor Immune Microenvironment
by Lisha Li, Zhen Yu, Xuehua Lu, Pei Yang, Shuxia Zhang, Qinghua Lin and Junyong Han
Curr. Issues Mol. Biol. 2025, 47(12), 991; https://doi.org/10.3390/cimb47120991 - 27 Nov 2025
Viewed by 389
Abstract
Background: Adhesion G protein-coupled receptor G6 (ADGRG6), also known as GPR126, has been implicated in several malignancies. However, its expression pattern, clinical significance, and mechanistic role in pancreatic adenocarcinoma (PAAD) remain unclear. Methods: We combined multi-omics analyses, tissue microarray immunohistochemistry, [...] Read more.
Background: Adhesion G protein-coupled receptor G6 (ADGRG6), also known as GPR126, has been implicated in several malignancies. However, its expression pattern, clinical significance, and mechanistic role in pancreatic adenocarcinoma (PAAD) remain unclear. Methods: We combined multi-omics analyses, tissue microarray immunohistochemistry, and a series of functional experiments, including 2D and 3D spheroid cultures, zebrafish xenografts, and murine tumor models—to investigate the expression, clinical significance, and mechanism of ADGRG6 in PAAD. The association between ADGRG6 expression and immune infiltration was assessed using TIMER and GEPIA databases, followed by mechanistic validation through ADGRG6 modulation in PAAD cell lines. Results: ADGRG6 was significantly overexpressed in PAAD and correlated with larger tumor size, higher grade, advanced TNM stage, and poor overall survival. Multivariate logistic regression confirmed that high ADGRG6 expression was independently associated with higher pathological grade. Functionally, ADGRG6 silencing markedly inhibited PAAD cell proliferation, migration, and invasion in both 2D and 3D cultures, as well as in zebrafish and nude mouse xenograft models. Integrated transcriptomic and immune analyses revealed that ADGRG6 expression positively correlated with mast cells, macrophages (M1/M2), Th2/Th17 subsets, and interferon–responsive neutrophils. Mechanistically, ADGRG6 silencing reduced STAT6 phosphorylation and GATA3 expression, consistent with the suppression of the NF-κB→STAT6→GATA3 axis. Conclusions: ADGRG6 functions as an oncogenic driver in PAAD, promoting tumor progression and fostering an immunosuppressive microenvironment via NF-κB/STAT6 signaling. These findings not only broaden the mechanistic understanding of ADGRG6 function but also suggest it as a promising target for therapeutic intervention in PAAD. Full article
Show Figures

Figure 1

19 pages, 1571 KB  
Review
From Spheroids to Tumor-on-a-Chip for Cancer Modeling and Therapeutic Testing
by Maria Veronica Lipreri, Marilina Tamara Totaro, Nicola Baldini and Sofia Avnet
Micromachines 2025, 16(12), 1343; https://doi.org/10.3390/mi16121343 - 27 Nov 2025
Cited by 1 | Viewed by 1078
Abstract
The high failure rate of anticancer drugs in clinical trials highlights the need for preclinical models that accurately reproduce the structural, biochemical, and mechanical complexity of human tumors. Conventional two-dimensional cultures and animal models often lack the physiological complexity required to predict clinical [...] Read more.
The high failure rate of anticancer drugs in clinical trials highlights the need for preclinical models that accurately reproduce the structural, biochemical, and mechanical complexity of human tumors. Conventional two-dimensional cultures and animal models often lack the physiological complexity required to predict clinical outcomes, driving the development of three-dimensional systems that better emulate the tumor microenvironment. Among these, microfluidic-based spheroid models have emerged as powerful tools for cancer research and drug screening. By integrating 3D spheroids with microfluidics, these platforms allow precise control of nutrient flow, oxygen gradients, shear stress, and interstitial pressure, while supporting co-culture with stromal, immune, and endothelial cells. Such systems enable the investigation of drug response, angiogenesis, metastasis, and immune interactions under dynamic and physiologically relevant conditions. This review summarizes recent advances in microfluidic spheroid models for cancer, covering both carcinomas and sarcomas, with an emphasis on device design, biomaterial integration, and translational validation. Key challenges remain, including technical complexity, scalability constraints, and the absence of standardized protocols. Overall, the merger of microfluidic technology with 3D spheroid culture provides a promising pathway toward predictive, ethical, and personalized preclinical testing, bridging the gap between in vitro modeling and clinical oncology. Full article
(This article belongs to the Special Issue Development of 3D Cancer Models in Microengineered Systems)
Show Figures

Figure 1

15 pages, 7506 KB  
Article
Pan-Cancer Analysis Reveals AEBP1-Collagen Co-Expression and Its Potential Role in CAF-Mediated Tumor Stiffness
by Shohei Sekiguchi, Akira Yorozu, Megumi Watanabe, Fumika Okazaki, Satoshi Ohwada, Eiichiro Yamamoto, Takeshi Niinuma, Hiroshi Kitajima, Kazuya Ishiguro, Mitsunobu Saito, Masahiro Kai, Masashi Idogawa, Kenichi Takano, Akihiro Miyazaki, Hiroshi Ohguro and Hiromu Suzuki
Int. J. Mol. Sci. 2025, 26(23), 11474; https://doi.org/10.3390/ijms262311474 - 27 Nov 2025
Viewed by 824
Abstract
Cancer-associated fibroblasts (CAFs) are critical components of the tumor microenvironment that promote cancer progression and immune evasion. Adipocyte enhancer-binding protein 1 gene (AEBP1), which encodes aortic carboxypeptidase-like protein (ACLP), has been implicated in tissue remodeling and fibrosis, yet its role in [...] Read more.
Cancer-associated fibroblasts (CAFs) are critical components of the tumor microenvironment that promote cancer progression and immune evasion. Adipocyte enhancer-binding protein 1 gene (AEBP1), which encodes aortic carboxypeptidase-like protein (ACLP), has been implicated in tissue remodeling and fibrosis, yet its role in CAF biology across cancers remains poorly understood. Here, we performed a pan-cancer transcriptomic analysis using The Cancer Genome Atlas (TCGA) and found that AEBP1 expression strongly correlates with expression of collagen family genes in the majority of solid tumors. Integration of single-cell RNA-sequencing datasets from breast and pancreatic cancers revealed that AEBP1 is predominantly expressed in CAFs, where it is co-expressed with collagens and CAF marker genes. Functional experiments using three-dimensional (3D) spheroids composed of oral squamous cell carcinoma (OSCC)-derived CAFs showed that AEBP1 knockdown significantly reduced spheroid stiffness without altering their morphology or size, indicating that ACLP contributes to the mechanical properties of tumor tissues. Together with earlier findings linking AEBP1/ACLP to reduced CD8+ T-cell infiltration, our results suggest that stromal AEBP1/ACLP enhances both extracellular matrix stiffness and immune suppression and highlights AEBP1/ACLP as a potential therapeutic target through which to remodel the tumor microenvironment and improve anti-tumor immunity. Full article
(This article belongs to the Special Issue Cancer-Associated Stromal Cells)
Show Figures

Figure 1

18 pages, 2737 KB  
Article
E-Cadherin Regulates HIF1-α In Vitro in Induced 3D Spheroid Models of Human Breast Cancer Through Both mTOR and Notch1 Signaling
by Yin Ye, Dollada Srisai and Sanford H. Barsky
Biomedicines 2025, 13(12), 2890; https://doi.org/10.3390/biomedicines13122890 - 26 Nov 2025
Viewed by 451
Abstract
Background: Both spontaneous and induced 3D spheroid models are among many in vitro models that recapitulate aspects of in vivo cancers. Although numerous studies have described the spatiotemporal relevance of these 3D models, there has been a paucity of studies investigating the [...] Read more.
Background: Both spontaneous and induced 3D spheroid models are among many in vitro models that recapitulate aspects of in vivo cancers. Although numerous studies have described the spatiotemporal relevance of these 3D models, there has been a paucity of studies investigating the signaling pathways that are activated during spheroidgenesis. Methods: Since in vitro 3D spheroidgenesis is thought to reflect at least some of the in vivo aspects of cancer biology (which undoubtedly involve cell adhesion, metabolism, and hypoxia-related pathways) and since we previously investigated these pathways in a model of spontaneous spheroidgenesis, this present study investigates these pathways in a model of induced spheroidgenesis with comparative studies involving a series of well-known E-cadherin-positive (MCF-7, HTB-126, HTB-27) and E-cadherin-negative (MDA-MB-468, MDA-MB-231, BT-549) human breast carcinoma cell lines. Results: Our findings demonstrate that during early induced spheroidgenesis, E-cadherin regulates hypoxia-inducible factor 1-alpha (HIF-1α) predominantly through PI3K/AKT/mTOR signaling and to a lesser extent through Notch1 signaling. Both the knockout of E-cadherin and calpain-mediated E-cadherin proteolysis result in a remarkable reduction in HIF-1α. Conclusions: 3D spheroid models recapitulate, in part, some of the in vivo stages of cancer progression, which include primary tumor clusters, lymphovascular emboli, and micrometastases, the signaling pathways present in these 3D spheroid models likely have relevance in vivo. Full article
(This article belongs to the Special Issue Molecular Research in Breast Cancer)
Show Figures

Graphical abstract

18 pages, 5178 KB  
Article
Properties of 3-Dimensional Cell Cultivation Matrices and Scaffolds in Magnetic Resonance Imaging at 3 Tesla
by Rebecca Wißmann, Petros Martirosian, Marina Danalache, Stefanie Elser, Jürgen Machann and Fritz Schick
J. Funct. Biomater. 2025, 16(12), 440; https://doi.org/10.3390/jfb16120440 - 26 Nov 2025
Viewed by 801
Abstract
Natural or synthetic scaffolds are essential for developing three-dimensional (3D) cell culture models, as they provide structural stability and accurately replicate the cellular microenvironment. When integrated into optimized setups, scaffold-supported cellular aggregates, such as spheroids, can be non-destructively characterized and monitored using 3T [...] Read more.
Natural or synthetic scaffolds are essential for developing three-dimensional (3D) cell culture models, as they provide structural stability and accurately replicate the cellular microenvironment. When integrated into optimized setups, scaffold-supported cellular aggregates, such as spheroids, can be non-destructively characterized and monitored using 3T Magnetic Resonance Imaging (MRI). However, a significant technical limitation is the presence of MR artifacts generated by scaffolds, which can severely obscure the visualization of the embedded spheroids. This study systematically evaluated the suitability of various scaffolds and matrices (including Matrigel®, fibrin glue, and several hydrogels) for MRI and MR spectroscopy (MRS). The materials were investigated both native and seeded with chondrosarcoma cells (SW1353). Our findings revealed considerable variability in MR compatibility across different materials. Specifically, fibrin glue proved unsuitable for MR applications due to substantial artifact generation that interfered with the visualization of cellular components. Furthermore, the results emphasize the importance of the observation period, as material degradation processes can introduce confounding factors in longitudinal MR studies. The choice of scaffold material is paramount for the successful analysis of 3D cell models via MRI. Careful selection is required, as the materials’ properties and temporal stability directly impact the interpretability of the acquired data. Full article
Show Figures

Figure 1

Back to TopTop