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

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (177)

Search Parameters:
Keywords = hypoxia heterogeneity

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
26 pages, 6895 KiB  
Article
Generation of Individualized, Standardized, and Electrically Synchronized Human Midbrain Organoids
by Sanae El Harane, Bahareh Nazari, Nadia El Harane, Manon Locatelli, Bochra Zidi, Stéphane Durual, Abderrahim Karmime, Florence Ravier, Adrien Roux, Luc Stoppini, Olivier Preynat-Seauve and Karl-Heinz Krause
Cells 2025, 14(15), 1211; https://doi.org/10.3390/cells14151211 - 6 Aug 2025
Abstract
Organoids allow to model healthy and diseased human tissues. and have applications in developmental biology, drug discovery, and cell therapy. Traditionally cultured in immersion/suspension, organoids face issues like lack of standardization, fusion, hypoxia-induced necrosis, continuous agitation, and high media volume requirements. To address [...] Read more.
Organoids allow to model healthy and diseased human tissues. and have applications in developmental biology, drug discovery, and cell therapy. Traditionally cultured in immersion/suspension, organoids face issues like lack of standardization, fusion, hypoxia-induced necrosis, continuous agitation, and high media volume requirements. To address these issues, we developed an air–liquid interface (ALi) technology for culturing organoids, termed AirLiwell. It uses non-adhesive microwells for generating and maintaining individualized organoids on an air–liquid interface. This method ensures high standardization, prevents organoid fusion, eliminates the need for agitation, simplifies media changes, reduces media volume, and is compatible with Good Manufacturing Practices. We compared the ALi method to standard immersion culture for midbrain organoids, detailing the process from human pluripotent stem cell (hPSC) culture to organoid maturation and analysis. Air–liquid interface organoids (3D-ALi) showed optimized size and shape standardization. RNA sequencing and immunostaining confirmed neural/dopaminergic specification. Single-cell RNA sequencing revealed that immersion organoids (3D-i) contained 16% fibroblast-like, 23% myeloid-like, and 61% neural cells (49% neurons), whereas 3D-ALi organoids comprised 99% neural cells (86% neurons). Functionally, 3D-ALi organoids showed a striking electrophysiological synchronization, unlike the heterogeneous activity of 3D-i organoids. This standardized organoid platform improves reproducibility and scalability, demonstrated here with midbrain organoids. The use of midbrain organoids is particularly relevant for neuroscience and neurodegenerative diseases, such as Parkinson’s disease, due to their high incidence, opening new perspectives in disease modeling and cell therapy. In addition to hPSC-derived organoids, the method’s versatility extends to cancer organoids and 3D cultures from primary human cells. Full article
(This article belongs to the Special Issue The Current Applications and Potential of Stem Cell-Derived Organoids)
Show Figures

Figure 1

18 pages, 3891 KiB  
Review
Navigating Brain Organoid Maturation: From Benchmarking Frameworks to Multimodal Bioengineering Strategies
by Jingxiu Huang, Yingli Zhu, Jiong Tang, Yang Liu, Ming Lu, Rongxin Zhang and Alfred Xuyang Sun
Biomolecules 2025, 15(8), 1118; https://doi.org/10.3390/biom15081118 - 4 Aug 2025
Viewed by 266
Abstract
Brain organoid technology has revolutionized in vitro modeling of human neurodevelopment and disease, providing unprecedented insights into cortical patterning, neural circuit assembly, and pathogenic mechanisms of neurological disorders. Critically, human brain organoids uniquely recapitulate human-specific developmental processes—such as the expansion of outer radial [...] Read more.
Brain organoid technology has revolutionized in vitro modeling of human neurodevelopment and disease, providing unprecedented insights into cortical patterning, neural circuit assembly, and pathogenic mechanisms of neurological disorders. Critically, human brain organoids uniquely recapitulate human-specific developmental processes—such as the expansion of outer radial glia and neuromelanin—that are absent in rodent models, making them indispensable for studying human brain evolution and dysfunction. However, a major bottleneck persists: Extended culture periods (≥6 months) are empirically required to achieve late-stage maturation markers like synaptic refinement, functional network plasticity, and gliogenesis. Yet prolonged conventional 3D culture exacerbates metabolic stress, hypoxia-induced necrosis, and microenvironmental instability, leading to asynchronous tissue maturation—electrophysiologically active superficial layers juxtaposed with degenerating cores. This immaturity/heterogeneity severely limits their utility in modeling adult-onset disorders (e.g., Alzheimer’s disease) and high-fidelity drug screening, as organoids fail to recapitulate postnatal transcriptional signatures or neurovascular interactions without bioengineering interventions. We summarize emerging strategies to decouple maturation milestones from rigid temporal frameworks, emphasizing the synergistic integration of chronological optimization (e.g., vascularized co-cultures) and active bioengineering accelerators (e.g., electrical stimulation and microfluidics). By bridging biological timelines with scalable engineering, this review charts a roadmap to generate translationally relevant, functionally mature brain organoids. Full article
Show Figures

Figure 1

23 pages, 1912 KiB  
Review
Utility of Multicellular Spheroids for Investigating Mechanisms of Chemoresistance in Triple-Negative Breast Cancer
by Keith N. Ncube, Iman van den Bout, Clarissa Willers, Chrisna Gouws and Werner Cordier
Int. J. Mol. Sci. 2025, 26(15), 7503; https://doi.org/10.3390/ijms26157503 - 3 Aug 2025
Viewed by 181
Abstract
Chemoresistance is a major challenge in the treatment of triple-negative breast cancer (TNBC). Multicellular spheroids are an attractive platform for investigating chemoresistance in TNBC, as they replicate the cues of the tumour microenvironment in vivo. We conducted a comprehensive literature search to summarise [...] Read more.
Chemoresistance is a major challenge in the treatment of triple-negative breast cancer (TNBC). Multicellular spheroids are an attractive platform for investigating chemoresistance in TNBC, as they replicate the cues of the tumour microenvironment in vivo. We conducted a comprehensive literature search to summarise the multifactorial and interlinked mechanisms driving chemoresistance in TNBC spheroids. These mechanisms include spatial heterogeneity, hypoxia, extracellular matrix remodelling, tumour–stroma crosstalk, drug efflux, apoptotic resistance, and cancer stem cell signalling. Strategies for overcoming chemoresistance in TNBC spheroids include nanocarrier systems to overcome spatial diffusion limitations, pathway inhibition, and targeting tumour–microenvironment interactions. Despite their advantages, some spheroid models face challenges such as low reproducibility, a lack of heterogeneity, variability in size and shape, limited vascularisation, and constraints in long-term culture. Advanced culturing platforms such as clinostat bioreactors allow for extended culture periods, enabling mature spheroid drug testing. Furthermore, advanced analytical techniques provide spatially resolved spheroid data. These multifactorial and interlinked mechanisms reflect the tumour microenvironment in vivo that spheroids recapitulate, rendering them valuable models for studying chemoresistance. The incorporation of stromal components and advanced analytical workflows will enhance the utility and translational relevance of spheroids as reliable preclinical models for drug discovery in TNBC. Full article
(This article belongs to the Special Issue Recent Advances in 3D Tumor Models for Cancer Research)
Show Figures

Graphical abstract

52 pages, 3227 KiB  
Review
Advances in Photodynamic Treatment of Precancerous and Cancerous Gynecological Diseases
by Polina Alekseeva, Vladimir Makarov, Kanamat Efendiev, Aida Gilyadova and Victor Loschenov
Cancers 2025, 17(15), 2421; https://doi.org/10.3390/cancers17152421 - 22 Jul 2025
Viewed by 406
Abstract
High mortality rates and poor quality of life result from the late-stage detection and frequent recurrence of gynecological neoplasms. Background/Objectives: The aim of this study was to conduct a systematic analysis of the energy parameters of photodynamic therapy (PDT) in the treatment [...] Read more.
High mortality rates and poor quality of life result from the late-stage detection and frequent recurrence of gynecological neoplasms. Background/Objectives: The aim of this study was to conduct a systematic analysis of the energy parameters of photodynamic therapy (PDT) in the treatment of cervical and vulvar lesions, with a focus on stimulating immune responses leading to human papillomavirus (HPV) eradication and lesion regression without adverse effects, such as thermal damage. Methods: A total of 46 peer-reviewed studies published between January 2010 and April 2024 were analyzed. These studies focused on PDT applications for cervical and vulvar lesions, sourced from Google Scholar, Scopus, and Web of Science. Results: Although PDT shows promise, significant limitations exist, such as insufficient consideration of individual tumor characteristics, restricted treatment depths, and the heterogeneous distribution and low selectivity of photosensitizer (PS) accumulation in tumors. Tumor hypoxia further reduces PDT’s effectiveness, and most studies overlook immune system activation, which is crucial for targeting HPV infections and improving antitumor responses. Conclusions: Advancing the research into PDT’s molecular and cellular mechanisms, optimizing the immune response stimulation, and improving the PS and delivery methods could enhance the safety and effectiveness of cervical and vulvar neoplasm treatments. The use of personalized PDT parameters may reduce the side effects and enhance the outcomes for patients suffering from gynecological diseases. Full article
(This article belongs to the Special Issue New Advances in the Diagnosis of Gynecological Cancer)
Show Figures

Figure 1

32 pages, 1319 KiB  
Review
Effects of Targeted Radionuclide Therapy on Cancer Cells Beyond the Ablative Radiation Dose
by Guillermina Ferro-Flores, Erika Azorín-Vega, Blanca Ocampo-García, Myrna Luna-Gutiérrez, Pedro Cruz-Nova and Laura Meléndez-Alafort
Int. J. Mol. Sci. 2025, 26(14), 6968; https://doi.org/10.3390/ijms26146968 - 20 Jul 2025
Viewed by 657
Abstract
Targeted radionuclide therapy (TRT) utilizes radiopharmaceuticals to deliver radiation directly to cancer cells while sparing healthy tissues. Beyond the absorbed dose of ablative radiation, TRT induces non-targeted effects (NTEs) that significantly enhance its therapeutic efficacy. These effects include radiation-induced bystander effects (RIBEs), abscopal [...] Read more.
Targeted radionuclide therapy (TRT) utilizes radiopharmaceuticals to deliver radiation directly to cancer cells while sparing healthy tissues. Beyond the absorbed dose of ablative radiation, TRT induces non-targeted effects (NTEs) that significantly enhance its therapeutic efficacy. These effects include radiation-induced bystander effects (RIBEs), abscopal effects (AEs), radiation-induced genomic instability (RIGI), and adaptive responses, which collectively influence the behavior of cancer cells and the tumor microenvironment (TME). TRT also modulates immune responses, promoting immune-mediated cell death and enhancing the efficacy of combination therapies, such as the use of immune checkpoint inhibitors. The molecular mechanisms underlying TRT involve DNA damage, oxidative stress, and apoptosis, with repair pathways like homologous recombination (HR) and non-homologous end joining (NHEJ) playing critical roles. However, challenges such as tumor heterogeneity, hypoxia, and radioresistance limit the effectiveness of this approach. Advances in theranostics, which integrate diagnostic imaging with TRT, have enabled personalized treatment approaches, while artificial intelligence and improved dosimetry offer potential for treatment optimization. Despite the significant survival benefits of TRT in prostate cancer and neuroendocrine tumors, 30–40% of patients remain unresponsive, which highlights the need for further research into molecular pathways, long-term effects, and combined therapies. This review outlines the dual mechanisms of TRT, direct toxicity and NTEs, and discusses strategies to enhance its efficacy and expand its use in oncology. Full article
(This article belongs to the Special Issue Targeted Therapy of Cancer: Innovative Drugs and Molecular Tools)
Show Figures

Figure 1

28 pages, 2091 KiB  
Review
Spatiotemporal Heterogeneity of Tumor Glucose Metabolism Reprogramming: From Single-Cell Mechanisms to Precision Interventions
by Xiaoxue Chai, Qian Tao and Lili Li
Int. J. Mol. Sci. 2025, 26(14), 6901; https://doi.org/10.3390/ijms26146901 - 18 Jul 2025
Viewed by 602
Abstract
Glucose metabolism reprogramming as a defining hallmark of cancer has become a pivotal frontier in oncology research. Recent technological advances in single-cell sequencing, spatial omics, and metabolic imaging have transformed the field from static bulk analyses to dynamic investigations of spatiotemporal heterogeneity at [...] Read more.
Glucose metabolism reprogramming as a defining hallmark of cancer has become a pivotal frontier in oncology research. Recent technological advances in single-cell sequencing, spatial omics, and metabolic imaging have transformed the field from static bulk analyses to dynamic investigations of spatiotemporal heterogeneity at a single-cell resolution. This review systematically summarizes the current knowledge on tumor glucose metabolism dynamics, discussing spatial heterogeneity and temporal evolution patterns, metabolic subpopulation interactions revealed by single-cell metabolomics, the glucose metabolism–epigenetics–immunology regulatory axis, and therapeutic strategies targeting metabolic vulnerabilities. Recent technological advances in single-cell sequencing and spatial omics have transformed our understanding of tumor glucose metabolism by providing high-resolution insights into metabolic heterogeneity and regulatory mechanisms, contrasting with classical bulk analyses. Spatiotemporal heterogeneity critically influences therapeutic outcomes by enabling tumor cells to adapt metabolically under selective pressures (e.g., hypoxia, nutrient deprivation), fostering treatment resistance and relapse. Deciphering these dynamics is essential for developing spatiotemporally targeted strategies that address intratumoral diversity and microenvironmental fluctuations. By integrating cutting-edge advances, this review deepens our understanding of tumor metabolic complexity and provides a conceptual framework for developing spatiotemporally precise interventions. Full article
Show Figures

Figure 1

47 pages, 2757 KiB  
Review
Influence of Hypoxia on Tumor Heterogeneity, DNA Repair, and Cancer Therapy: From Molecular Insights to Therapeutic Strategies
by Dominika Kunachowicz, Paulina Tomecka, Mikołaj Sędzik, Jarosław Kalinin, Jacek Kuźnicki and Nina Rembiałkowska
Cells 2025, 14(14), 1057; https://doi.org/10.3390/cells14141057 - 10 Jul 2025
Viewed by 744
Abstract
Hypoxia, characterized by a reduction in tissue oxygen levels, is a hallmark of many solid tumors and affects a range of cellular processes, including DNA repair. In low-oxygen conditions, cancer cells often suppress key DNA repair pathways such as homologous recombination (HR), leading [...] Read more.
Hypoxia, characterized by a reduction in tissue oxygen levels, is a hallmark of many solid tumors and affects a range of cellular processes, including DNA repair. In low-oxygen conditions, cancer cells often suppress key DNA repair pathways such as homologous recombination (HR), leading to the accumulation of DNA damage and increased genomic instability. These changes not only drive tumor progression but also contribute to resistance against conventional therapies. Hypoxia significantly reduces the effectiveness of oxygen-dependent treatments, including radiotherapy and many chemotherapeutic agents. To address this limitation, bioreductive drugs have been developed that become selectively activated in hypoxic environments, providing targeted cytotoxic effects within oxygen-deprived tumor regions. Additionally, the rapid growth of tumors often results in disorganized and inefficient vasculature, further impairing the delivery of oxygen and therapeutic agents. This review explores the molecular mechanisms by which hypoxia disrupts DNA repair and contributes to treatment resistance. It also presents emerging therapeutic strategies aimed at targeting the hypoxic tumor microenvironment to improve treatment efficacy and patient outcomes. Full article
(This article belongs to the Section Cell Microenvironment)
Show Figures

Figure 1

29 pages, 909 KiB  
Review
Unraveling the Role of Tumor-Infiltrating Immune Cells in Head and Neck Squamous Cell Carcinoma: Implications for Antitumor Immune Responses and Immunotherapy
by Paula Constanza Arriola Benítez, Mariel Fusco, Ricardo Amorin, Carlos Rafael Picón, Flavia Piccioni, Lucia Victoria, Manglio Miguel Rizzo and Mariana Malvicini
Int. J. Mol. Sci. 2025, 26(13), 6337; https://doi.org/10.3390/ijms26136337 - 30 Jun 2025
Viewed by 936
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a highly heterogeneous malignancy characterized by a complex tumor microenvironment (TME) that plays a critical role in disease progression and therapeutic resistance. Tumor-infiltrating immune cells, including T lymphocytes, macrophages, dendritic cells, and myeloid-derived suppressor cells, [...] Read more.
Head and neck squamous cell carcinoma (HNSCC) is a highly heterogeneous malignancy characterized by a complex tumor microenvironment (TME) that plays a critical role in disease progression and therapeutic resistance. Tumor-infiltrating immune cells, including T lymphocytes, macrophages, dendritic cells, and myeloid-derived suppressor cells, exhibit dual functions, either promoting or suppressing tumor growth depending on their phenotype and interactions within the TME. The presence of immune evasion mechanisms, such as the loss of human leukocyte antigen (HLA) expression, upregulation of immune checkpoint molecules, and metabolic reprogramming (hypoxia-induced glycolysis and lactate accumulation), further contributes to immune suppression and poor treatment responses. While immune checkpoint inhibitors (ICIs) have revolutionized the treatment of recurrent/metastatic HNSCC, response rates remain highly variable, underscoring the need for biomarker-driven patient selection and combinatorial therapeutic strategies. This review provides a comprehensive analysis of the role of immune cells in the TME of HNSCC, discusses the mechanisms underlying immune escape, and explores emerging immunotherapeutic and epigenetic-targeting approaches aimed at enhancing antitumor immune responses and improving clinical outcomes. Full article
Show Figures

Figure 1

34 pages, 1793 KiB  
Review
Deciphering Radiotherapy Resistance: A Proteomic Perspective
by Davide Perico and Pierluigi Mauri
Proteomes 2025, 13(2), 25; https://doi.org/10.3390/proteomes13020025 - 16 Jun 2025
Viewed by 685
Abstract
Radiotherapy resistance represents a critical aspect of cancer treatment, and molecular characterization is needed to explore the pathways and mechanisms involved. DNA repair, hypoxia, metabolic reprogramming, apoptosis, tumor microenvironment modulation, and activation of cancer stem cells are the primary mechanisms that regulate radioresistance, [...] Read more.
Radiotherapy resistance represents a critical aspect of cancer treatment, and molecular characterization is needed to explore the pathways and mechanisms involved. DNA repair, hypoxia, metabolic reprogramming, apoptosis, tumor microenvironment modulation, and activation of cancer stem cells are the primary mechanisms that regulate radioresistance, and understanding their complex interactions is essential for planning the correct therapeutic strategy. Proteomics has emerged as a key approach in precision medicine to study tumor heterogeneity and treatment response in cancer patients. The integration of mass spectrometry-based techniques with bioinformatics has enabled high-throughput, quantitative analyses to identify biomarkers, pathways, and new potential therapeutic targets. This review highlights recent advances in proteomic technologies and their application in identifying biomarkers predictive of radiosensitivity and radioresistance in different tumors, including head and neck, breast, lung, and prostate cancers. Sample variability, data interpretation, and the translation of findings into clinical practice remain challenging elements of proteomics. However, technological advancements support its application in a wide range of topics, allowing a comprehensive approach to radiobiology, which helps overcome radiation resistance. Ultimately, incorporating proteomics into the radiotherapy workflow offers significant potential for enhancing treatment efficacy, minimizing toxicity, and guiding precision oncology strategies. Full article
(This article belongs to the Special Issue Clinical Proteomics: Fourth Edition)
Show Figures

Graphical abstract

39 pages, 778 KiB  
Review
Epigenetic Drivers of Atrial Fibrillation: Mechanisms, Biomarkers, and Therapeutic Targets
by Paschalis Karakasis, Panagiotis Theofilis, Nikias Milaras, Panayotis K. Vlachakis, Dimitrios Patoulias, Theodoros Karamitsos, Antonios P. Antoniadis and Nikolaos Fragakis
Int. J. Mol. Sci. 2025, 26(11), 5253; https://doi.org/10.3390/ijms26115253 - 29 May 2025
Cited by 3 | Viewed by 859
Abstract
Atrial fibrillation (AF) is the most prevalent sustained arrhythmia, associated with significant morbidity, mortality, and healthcare burdens. Despite therapeutic advances, recurrence rates remain high, particularly in persistent AF, underscoring the need for deeper mechanistic insight. Epigenetic regulation—comprising DNA methylation, histone modifications, chromatin remodeling, [...] Read more.
Atrial fibrillation (AF) is the most prevalent sustained arrhythmia, associated with significant morbidity, mortality, and healthcare burdens. Despite therapeutic advances, recurrence rates remain high, particularly in persistent AF, underscoring the need for deeper mechanistic insight. Epigenetic regulation—comprising DNA methylation, histone modifications, chromatin remodeling, RNA methylation, and non-coding RNAs—has emerged as a key contributor to the structural, electrical, and inflammatory remodeling underlying AF. These mechanisms operate at the interface of genetic susceptibility and environmental exposure, offering a dynamic framework for understanding disease progression. Systemic stressors such as aging, obesity, diabetes, hypertension, hypoxia, and alcohol have been shown to induce epigenetic reprogramming in atrial tissue, further promoting atrial cardiomyopathy and arrhythmogenesis. Additionally, circulating epigenetic markers, particularly microRNAs, are being investigated for their potential in AF diagnosis, risk stratification, and therapeutic monitoring. Therapeutic strategies targeting epigenetic pathways—ranging from histone deacetylase inhibitors and miRNA-based therapeutics to CRISPR/dCas9-mediated epigenome editing—are under investigation. Additionally, sodium-glucose cotransporter 2 inhibitors may indirectly influence epigenetic programs and miRNA expression relevant to atrial remodeling. While promising, these approaches require further validation in terms of safety, delivery specificity, and long-term efficacy. High-resolution epigenomic mapping and integrative multi-omic approaches may enhance understanding of AF heterogeneity and enable personalized treatment strategies. This review provides an integrated appraisal of epigenetic mechanisms in AF and outlines their emerging diagnostic and therapeutic relevance. Full article
Show Figures

Figure 1

16 pages, 1934 KiB  
Review
Pathophysiology in Systemic Sclerosis: Current Insights and Future Perspectives
by Suzan Al-Gburi, Pia Moinzadeh and Thomas Krieg
Sclerosis 2025, 3(2), 17; https://doi.org/10.3390/sclerosis3020017 - 27 May 2025
Viewed by 1215
Abstract
Background: Systemic sclerosis (SSc) is a rare connective tissue disease characterized by vasculopathy, autoimmunity, and fibrosis. Due to its low prevalence and heterogeneous clinical presentation, early diagnosis remains challenging, often delaying appropriate treatment. The disease progresses from microvascular dysfunction, manifesting as Raynaud’s phenomenon, [...] Read more.
Background: Systemic sclerosis (SSc) is a rare connective tissue disease characterized by vasculopathy, autoimmunity, and fibrosis. Due to its low prevalence and heterogeneous clinical presentation, early diagnosis remains challenging, often delaying appropriate treatment. The disease progresses from microvascular dysfunction, manifesting as Raynaud’s phenomenon, to systemic fibrosis affecting multiple organs, including the lungs, gastrointestinal tract, heart, and kidneys. There have been considerable advancements in understanding the pathophysiology of the disease during the last few years and this has already resulted in the improvement of the therapeutic approaches used to control organ-specific manifestations. However, the underlying cause of the disease still remains incompletely elucidated. Methods: Here, we summarize the current knowledge on the SSc pathogenesis. Results: The pathophysiology involves an interplay of chronic inflammation, impaired vascular function, and excessive extracellular matrix deposition, leading to progressive organ damage. Endothelial dysfunction in SSc is driven by immune-mediated injury, oxidative stress, and the imbalance of vasoconstrictors and vasodilators, leading to capillary loss and chronic hypoxia. Autoantibodies against endothelial cells or other toxic factors induce apoptosis and impair angiogenesis, further exacerbating vascular damage. Despite increased angiogenic factor levels, capillary repair mechanisms are defective, resulting in progressive ischemic damage. Dysregulated immune responses involving Th2 cytokines, B cells, and macrophages contribute to fibroblast activation and excessive collagen deposition. Transforming growth factor-beta (TGF-β) plays a central role in fibrotic progression, while fibroblasts resist apoptosis, perpetuating tissue scarring. The extracellular matrix in SSc is abnormally stiff, reinforcing fibroblast activation and creating a self-perpetuating fibrotic cycle. Conclusions: Advances in molecular and cellular understanding have facilitated targeted therapies, yet effective disease-modifying treatments remain limited. Future research should focus on precision medicine approaches, integrating biomarkers and novel therapeutics to improve patient outcomes. Full article
(This article belongs to the Special Issue Recent Advances in Understanding Systemic Sclerosis)
Show Figures

Figure 1

35 pages, 2316 KiB  
Review
Modeling Tumor Microenvironment Complexity In Vitro: Spheroids as Physiologically Relevant Tumor Models and Strategies for Their Analysis
by Shrey Shah and Gerard G. M. D’Souza
Cells 2025, 14(10), 732; https://doi.org/10.3390/cells14100732 - 17 May 2025
Cited by 1 | Viewed by 1329
Abstract
Drug delivery to solid tumors is challenged by multiple physiological barriers arising from the tumor microenvironment, including dense extracellular matrix, cellular heterogeneity, hypoxic gradients, and elevated interstitial fluid pressure. These features hinder the uniform distribution and accumulation of therapeutics, reducing treatment efficacy. Despite [...] Read more.
Drug delivery to solid tumors is challenged by multiple physiological barriers arising from the tumor microenvironment, including dense extracellular matrix, cellular heterogeneity, hypoxic gradients, and elevated interstitial fluid pressure. These features hinder the uniform distribution and accumulation of therapeutics, reducing treatment efficacy. Despite their widespread use, conventional two-dimensional monolayer cultures fail to reproduce these complexities, contributing to the poor translational predictability of many preclinical candidates. Three-dimensional multicellular tumor spheroids have emerged as more representative in vitro models that capture essential features of tumor architecture, stromal interactions, and microenvironmental resistance mechanisms. Spheroids exhibit spatially organized regions of proliferation, quiescence, and hypoxia, and can incorporate non-tumor cells to mimic tumor–stroma crosstalk. Advances in spheroid analysis now enable detailed evaluation of drug penetration, cellular migration, cytotoxic response, and molecular gradients using techniques such as optical and confocal imaging, large-particle flow cytometry, biochemical viability assays, and microfluidic integration. By combining physiological relevance with analytical accessibility, spheroid models support mechanistic studies of drug transport and efficacy under tumor-like conditions. Their adoption into routine preclinical workflows has the potential to improve translational accuracy while reducing reliance on animal models. Full article
(This article belongs to the Special Issue 3D Cultures and Organ-on-a-Chip in Cell and Tissue Cultures)
Show Figures

Graphical abstract

22 pages, 5751 KiB  
Article
Targeting Aggressive Prostate Carcinoma Cells with Mesothelin-CAR-T Cells
by Apolline de Testas de Folmont, Angèle Fauvel, Francis Vacherot, Pascale Soyeux, Abdérémane Abdou, Salem Chouaib and Stéphane Terry
Biomedicines 2025, 13(5), 1215; https://doi.org/10.3390/biomedicines13051215 - 16 May 2025
Viewed by 680
Abstract
Background: Advancing chimeric antigen receptor (CAR) T cell therapy for solid tumors remains a major challenge in cancer immunotherapy. Prostate cancer (PCa), particularly in its aggressive forms, may be a suitable target for CAR-T therapy given the range of associated tumor antigens. [...] Read more.
Background: Advancing chimeric antigen receptor (CAR) T cell therapy for solid tumors remains a major challenge in cancer immunotherapy. Prostate cancer (PCa), particularly in its aggressive forms, may be a suitable target for CAR-T therapy given the range of associated tumor antigens. However, due to the high plasticity and heterogeneity of aggressive PCa and the complexity of the tumor environment, there is a need to broaden the repertoire of targetable antigens and deepen our understanding of CAR-T behavior in stressed microenvironmental conditions. Growing evidence supports mesothelin as a promising cancer-associated marker and a compelling target for CAR-T cell approaches in solid tumors. Objectives and Methods: Here, we employed gene expression datasets to investigate mesothelin expression in both primary and metastatic PCa tumors. Additionally, we evaluated mesothelin expression across various preclinical PCa models and assessed the therapeutic efficacy of second-generation mesothelin-targeted CAR-T (meso-CAR-T) cells under both normoxic and hypoxic conditions, with hypoxia as a representative tumor-associated stress condition. Results: Our results revealed a significant enrichment of mesothelin in 3–10% of metastatic prostate tumors, contrasting with its minimal expression in primary tumors. In line with these findings, we observed increased mesothelin expression in an aggressive variant of the 22Rv1 cell line, which displayed an epithelial–mesenchymal plasticity (EMP) phenotype. Meso-CAR-T cells demonstrated potent cytotoxicity and remarkable selectivity toward these carcinoma cells under both severe hypoxia (1% O2) or normoxia (21% O2), highlighting their ability to withstand metabolic stress within the tumor microenvironment. Conclusions: Our study underscores the potential of meso-CAR-T cells as a promising strategy for targeting specific subtypes of metastatic prostate cancer. Full article
(This article belongs to the Special Issue The Development of Cancer Immunotherapy)
Show Figures

Figure 1

19 pages, 9021 KiB  
Systematic Review
The Association of Placental Grading with Perinatal Outcomes: A Systematic Review and Meta-Analysis
by Antonios Siargkas, Christina Pachi, Meletios P. Nigdelis, Sofoklis Stavros, Ekaterini Domali, Apostolos Mamopoulos, Ioannis Tsakiridis and Themistoklis Dagklis
Diagnostics 2025, 15(10), 1264; https://doi.org/10.3390/diagnostics15101264 - 15 May 2025
Cited by 1 | Viewed by 802
Abstract
Objective: Premature placental calcification (PPC) has been implicated in adverse perinatal outcomes, yet its clinical significance remains controversial. This meta-analysis aimed to quantitatively synthesize current data on the association between PPC, defined as grade 3 placental calcification before 36+6 weeks of [...] Read more.
Objective: Premature placental calcification (PPC) has been implicated in adverse perinatal outcomes, yet its clinical significance remains controversial. This meta-analysis aimed to quantitatively synthesize current data on the association between PPC, defined as grade 3 placental calcification before 36+6 weeks of gestation and adverse perinatal outcomes. Data Sources: A systematic search was conducted in MEDLINE, Scopus and The Cochrane Library from inception until 11 March 2025, to identify eligible studies. Study Eligibility Criteria: Observational studies including singleton pregnancies with PPC diagnosed via ultrasonography between 28+0 and 36+6 weeks of gestation and comparing them with pregnancies with Grannum grade 0, 1, or 2 placentas were considered eligible. Methods: Study quality was assessed using the Newcastle−Ottawa Scale, and the risk of bias was evaluated with the Quality In Prognosis Studies tool. The primary outcomes were small-for-gestational-age (SGA) neonates and preeclampsia. Heterogeneity was assessed using Cochran’s Q test and the I2 statistic. Meta-analyses were conducted using a random-effects model, with outcomes reported as relative risk (RR) or mean difference (MD) with 95% confidence intervals (CIs). Results: In total, nine cohort studies were included. PPC was associated with an increased risk of SGA (RR, 1.99; 95% CI, 1.46−2.70), preeclampsia (RR, 5.27; 95% CI, 2.24−12.40), fetal growth restriction (RR, 2.31; 95% CI, 1.30−4.09), preterm delivery (RR, 2.11; 95% CI, 1.00−4.45), suspected fetal hypoxia (RR, 1.71; 95% CI, 1.13–2.56), low 5 min Apgar score (RR, 2.28; 95% CI, 1.50−3.44) and neonatal intensive care unit admission (RR, 1.80; 95% CI, 1.02−3.18). No significant associations were found with fetal or neonatal death (RR, 2.75; 95% CI, 0.87−8.71), cesarean delivery (RR, 1.26; 95% CI, 0.90−1.78), gestational diabetes mellitus (RR, 1.17; 95% CI, 0.81−1.70), neonatal resuscitation (RR, 1.04; 95% CI, 0.92−1.16), birthweight (MD, −187.46 g; 95% CI, −413.14 to +38.21), or gestational age at birth (MD, −0.62 weeks; 95% CI, −1.36 to +0.11). A sensitivity analysis excluding high-risk-of-bias studies yielded consistent results. Conclusions: PPC is associated with several adverse perinatal outcomes, including SGA and preeclampsia. While the clinical significance of placental grading has remained limited in recent years, this study has shown that PPC may serve as an early indicator of placental insufficiency, warranting enhanced fetal surveillance and risk assessment in affected pregnancies. Further research is needed to refine its prognostic utility and integration into obstetric practice. Full article
Show Figures

Figure 1

22 pages, 6981 KiB  
Systematic Review
The Incidence of Adverse Events in Adults Undergoing Procedural Sedation with Propofol Administered by Non-Anesthetists: A Systematic Review and Meta-Analysis
by Flavia Pigò, Matteo Gottin and Rita Conigliaro
Diagnostics 2025, 15(10), 1234; https://doi.org/10.3390/diagnostics15101234 - 14 May 2025
Viewed by 698
Abstract
Background/Objectives: The administration of propofol without an anesthesiologist (NAAP) during endoscopic procedures is generally considered safe. However, the available data remain limited and fragmented due to legal constraints. This systematic review and meta-analysis aimed to evaluate the incidence of adverse events in adults [...] Read more.
Background/Objectives: The administration of propofol without an anesthesiologist (NAAP) during endoscopic procedures is generally considered safe. However, the available data remain limited and fragmented due to legal constraints. This systematic review and meta-analysis aimed to evaluate the incidence of adverse events in adults undergoing procedural sedation with NAAP. Methods: A comprehensive search was conducted in three electronic databases (MEDLINE, EMBASE, and the Cochrane Library) for studies published between 2010 and 2023. Eligible studies included randomized controlled trials and observational studies that reported predefined adverse events in adult patients receiving NAAP for procedural sedation. The analysis encompassed various types of endoscopic procedures and sedation protocols, including both balanced sedation and propofol monotherapy. Clinical heterogeneity was assessed by comparing patient characteristics, sedation methods, and outcome measures across studies. A random effects model was used for the meta-analysis, with results presented as estimated incidence rates. Subgroup analyses were conducted based on the hypoxia severity, sedation approach, and procedure type. Results: The search yielded 2963 records, of which 73 studies met the inclusion criteria, covering a total of 967,238 procedural sedations. Hypoxia was the most frequently reported adverse event, occurring in 40‰ of cases, followed by hypotension (38‰) and bradycardia (9‰). Severe adverse events requiring emergency intervention were rare, with an incidence of 0.12‰. The subgroup analysis indicated a low occurrence (6‰) of severe desaturation (SpO2 < 80%) and no significant differences in adverse event rates between balanced propofol sedation and propofol-only sedation. However, advanced endoscopic procedures (EUS, ERCP, PEG, enteroscopy, EMR/ESD) were associated with a higher risk of hypoxia (10% vs. 26‰; p < 0.00001) and major complications (3.1‰ vs. 0.1‰; p = 0.015) compared to diagnostic procedures. Conclusions: NAAP-based procedural sedation appears to be generally safe. While the minor adverse event rates vary depending on the sedation regimen and procedure type, major complications remain exceptionally rare. Full article
(This article belongs to the Section Clinical Diagnosis and Prognosis)
Show Figures

Figure 1

Back to TopTop