error_outline You can access the new MDPI.com website here. Explore and share your feedback with us.
 
 
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
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
remove_circle_outline
remove_circle_outline

Search Results (9,341)

Search Parameters:
Keywords = microenvironment tumor

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 861 KB  
Review
Emerging Oncogenic and Immunoregulatory Roles of BST2 in Human Cancers
by Chohee Kim, Seoyoon Choi and Jong-Whi Park
Biomedicines 2026, 14(1), 131; https://doi.org/10.3390/biomedicines14010131 (registering DOI) - 8 Jan 2026
Abstract
BST2 has emerged as a multifunctional molecule that bridges antiviral defense, membrane architecture, and tumor immunity. Originally characterized as an interferon-inducible restriction factor that tethers virions to the plasma membrane, BST2 is now recognized as an oncogenic driver and immunoregulatory hub in diverse [...] Read more.
BST2 has emerged as a multifunctional molecule that bridges antiviral defense, membrane architecture, and tumor immunity. Originally characterized as an interferon-inducible restriction factor that tethers virions to the plasma membrane, BST2 is now recognized as an oncogenic driver and immunoregulatory hub in diverse malignancies. In cancer, BST2 expression is frequently upregulated through promoter hypomethylation and transcriptional activation. Functionally, BST2 promotes proliferation, epithelial–mesenchymal transition, anoikis resistance, and chemoresistance, whereas its loss sensitizes tumor cells to proteotoxic and metabolic stresses. Beyond tumor cells, BST2 modulates the tumor microenvironment by promoting M2 macrophage infiltration, dendritic cell exhaustion, and natural killer (NK)-cell resistance, thereby contributing to immune evasion. Elevated BST2 expression correlates with poor prognosis in glioblastoma, breast, nasopharyngeal, and pancreatic cancers, and it serves as a circulating biomarker within small extracellular vesicles. In conclusion, BST2 is a dual-function molecule that integrates oncogenic signaling and immune regulation, making it an attractive diagnostic and therapeutic target for hematological and solid tumors. Full article
(This article belongs to the Special Issue Drug Resistance and Tumor Microenvironment in Human Cancers)
21 pages, 1832 KB  
Article
Nucleosome Clustering as a Biomarker and Mechanistic Switch for Reprogramming Cells
by Zhaoyuan Xu, Yinzhi Xu, Baiyan Li, Lidan You, Jing Liu and Hiroki Yokota
Cells 2026, 15(2), 113; https://doi.org/10.3390/cells15020113 - 8 Jan 2026
Abstract
Chromatin architecture is highly dynamic, undergoing nanoscale rearrangements throughout the cell cycle and in response to environmental cues. In this study, we employed high-resolution stochastic optical reconstruction microscopy (STORM) to visualize chromatin organization and cellular plasticity at the nanoscale in two osteosarcoma cell [...] Read more.
Chromatin architecture is highly dynamic, undergoing nanoscale rearrangements throughout the cell cycle and in response to environmental cues. In this study, we employed high-resolution stochastic optical reconstruction microscopy (STORM) to visualize chromatin organization and cellular plasticity at the nanoscale in two osteosarcoma cell lines, U2OS and MG63. To promote a tumor-suppressive bone microenvironment, we applied three biophysical modalities, namely mechanical vibration, electrical stimulation, and optical pulses, each previously linked to altered tumor behavior by reprogramming cells and generating induced tumor-suppressing (iTS) cells. These stimuli enlarged nuclear size and disrupted nuclear envelope integrity, as revealed by increased surface roughness. Critically, all three modalities transiently scattered nucleosome clusters, indicating chromatin decondensation as a hallmark of iTS cell generation. iTS cells exhibited elevated expression of histone demethylases lysine demethylase 3A (KDM3A) and lysine demethylase 4 (KDM4), accompanied by reduced levels of trimethylated histone H3 lysine 9 (H3K9me3). Consistently, pharmacological agents—Trichostatin A as a histone deacetylase inhibitor and chaetocin as a histone methyltransferase inhibitor—induced nucleosome scattering and converted U2OS cells into iTS cells, whose conditioned media exerted tumor-suppressive effects. Our findings highlight nucleosome clustering as a key epigenetic feature responsive to both biophysical and chemical cues, underscoring its role in microscale chromatin remodeling and reprogramming of the tumor microenvironment. Full article
(This article belongs to the Section Cellular Biophysics)
15 pages, 1216 KB  
Review
Autophagy Modulates Immunogenic Cell Death in Cancer
by Maiko Matsushita and Miyu Moriwaki
Cancers 2026, 18(2), 205; https://doi.org/10.3390/cancers18020205 - 8 Jan 2026
Abstract
Immunogenic cell death (ICD) is a subtype of regulated cell death characterized by the spatiotemporally coordinated emission of damage-associated molecular patterns (DAMPs), such as calreticulin (CALR), ATP, and high-mobility group box-1 (HMGB1), which collectively prime tumor-specific T-cell responses. Autophagy, a lysosome-dependent catabolic process, [...] Read more.
Immunogenic cell death (ICD) is a subtype of regulated cell death characterized by the spatiotemporally coordinated emission of damage-associated molecular patterns (DAMPs), such as calreticulin (CALR), ATP, and high-mobility group box-1 (HMGB1), which collectively prime tumor-specific T-cell responses. Autophagy, a lysosome-dependent catabolic process, is increasingly recognized as a key modifier of antitumor immunity and the tumor microenvironment (TME). In preclinical models, autophagy can not only promote ICD by sustaining endoplasmic reticulum (ER) stress, eukaryotic translation initiation factor-2α (eIF2α) phosphorylation, and secretory pathways, but it can also limit ICD by degrading DAMPs, antigenic cargo, and major histocompatibility complex (MHC) molecules. The net outcome is highly context-dependent and determined by the tumor type, the nature and intensity of the stress, and the level at which autophagy is modulated. Herein, we summarize how autophagy affects the three canonical ICD-associated DAMPs, highlight solid-tumor models in which autophagy supports ICD, and contrast them with systems wherein autophagy inhibition is required for immunogenicity. We then focus on hematological malignancies, especially multiple myeloma, where recent reports implicate the autophagy-related protein GABARAP in bortezomib-induced ICD. Finally, we discuss the translational implications, including rational combinations of autophagy modulators with ICD-inducing chemotherapies, targeted drugs, and cellular immunotherapies, and outline the remaining challenges for safely harnessing the autophagy–ICD axis in the clinical setting. Full article
(This article belongs to the Special Issue Autophagy and Apoptosis in Cancer Progression)
Show Figures

Figure 1

24 pages, 1126 KB  
Review
From Orange to Oncology: Anti-Inflammatory and Anti-Cancer Mechanisms of Sinensetin
by Dong Joon Kim, Songyeon Ahn, Xiaomeng Xie, Yeon-Sun Seong and Yong Weon Yi
Cells 2026, 15(2), 110; https://doi.org/10.3390/cells15020110 - 8 Jan 2026
Abstract
Sinensetin, a polymethoxylated flavone abundant in citrus fruits, has been recognized for its broad biological activities and wide use in traditional medicine around the world. Emerging clinical evidence from flavonoid-enriched orange juice interventions indicates antioxidant and anti-inflammatory effects, aligning with extensive preclinical data. [...] Read more.
Sinensetin, a polymethoxylated flavone abundant in citrus fruits, has been recognized for its broad biological activities and wide use in traditional medicine around the world. Emerging clinical evidence from flavonoid-enriched orange juice interventions indicates antioxidant and anti-inflammatory effects, aligning with extensive preclinical data. In this review, we explored in vitro and in vivo findings on the anti-inflammatory and anticancer actions of sinensetin and delineated the underlying cellular pathways, especially in terms of proposed targets for sinensetin. In inflammatory settings, sinensetin attenuates NF-κB activation, lowers pro-inflammatory cytokines (e.g., TNF-α, IL-6), and enhances antioxidant defenses, supporting its reported antioxidant, anti-bacterial, anti-viral, and anti-obesity properties. Across multiple tumor models, sinensetin suppresses oncogenic signaling—including β-catenin, PI3K/AKT, VEGF, NRF2, P53, and MKK6—concomitant with reduced proliferation, migration, and survival signaling. We further discuss emerging immunological effects, including modulation of innate immune cell activation and cytokine production, which may contribute to tumor microenvironment reprogramming and inflammation resolution. Together, these mechanistic insights position sinensetin as a promising lead for chemopreventive and adjunct therapeutic strategies. Our efforts aim to provide insights into the future translational development and clinical evaluation of sinensetin and its derivatives. Full article
Show Figures

Figure 1

17 pages, 20305 KB  
Article
Transcriptomic Analysis Identifies Acrolein Exposure-Related Pathways and Constructs a Prognostic Model in Oral Squamous Cell Carcinoma
by Yiting Feng, Lijuan Lou and Liangliang Ren
Int. J. Mol. Sci. 2026, 27(2), 632; https://doi.org/10.3390/ijms27020632 - 8 Jan 2026
Abstract
Acrolein, a highly reactive environmental toxicant widely present in urban air and tobacco smoke, has been implicated in the development of multiple malignancies. In oral tissues, chronic acrolein exposure induces oxidative stress, inflammation, and genetic mutations, all of which are closely linked to [...] Read more.
Acrolein, a highly reactive environmental toxicant widely present in urban air and tobacco smoke, has been implicated in the development of multiple malignancies. In oral tissues, chronic acrolein exposure induces oxidative stress, inflammation, and genetic mutations, all of which are closely linked to the development of oral squamous cell carcinoma (OSCC). Although accumulating evidence indicates a strong association between acrolein exposure and OSCC, its prognostic significance remains poorly understood. In this study, we analyzed transcriptome data to identify differentially expressed genes (DEGs) between tumor and adjacent normal tissues, and screened acrolein-related candidates by intersecting DEGs with previously identified acrolein-associated gene sets. Functional alterations of these genes were assessed using Gene Set Variation Analysis (GSVA), and a protein–protein interaction (PPI) network was constructed to identify key regulatory genes. A prognostic model was developed using Support Vector Machine–Recursive Feature Elimination (SVM-RFE) combined with LASSO-Cox regression and validated in an independent external cohort. Among the acrolein-related DEGs, four key genes (PLK1, AURKA, CTLA4, and PPARG) were ultimately selected for model construction. Kaplan–Meier analysis showed significantly worse overall survival in the high-risk group (p < 0.0001). Receiver operating characteristic (ROC) curve analysis further confirmed the strong predictive performance of the model, with area under the curve (AUC) values of 0.72 at 1 year, 0.72 at 3 years, and 0.75 at 5 years. Furthermore, the high risk score was significantly correlated with a ‘cold’ immune microenviroment, suggesting that acrolein-related genes may modulate the tumor immune microenvironment. Collectively, these findings highlight the role of acrolein in OSCC progression, suggesting the importance of reducing acrolein exposure for cancer prevention and public health, and call for increased attention to the relationship between environmental toxicants and disease initiation, providing a scientific basis for public health interventions and cancer prevention strategies. Full article
(This article belongs to the Special Issue Environmental Pollutants Exposure and Toxicity)
Show Figures

Figure 1

22 pages, 9895 KB  
Article
Targeting Lung Cancer Cell Motility Using Microbeam Radiation Therapy
by Ömer Dağkazanlı, Aleksandra Čolić, Rainer Lindner, Stefan Bartzsch, Stephanie E. Combs, Thomas E. Schmid and Marina Santiago Franco
Cells 2026, 15(2), 107; https://doi.org/10.3390/cells15020107 - 7 Jan 2026
Abstract
Radiotherapy (RT) is currently among the standard treatments for lung cancer. However, in vitro studies have revealed that irradiation can increase lung cancer cell motility. This way, RT could potentially enhance the malignancy of solid tumors post-treatment, promoting metastasis. Therefore, there is a [...] Read more.
Radiotherapy (RT) is currently among the standard treatments for lung cancer. However, in vitro studies have revealed that irradiation can increase lung cancer cell motility. This way, RT could potentially enhance the malignancy of solid tumors post-treatment, promoting metastasis. Therefore, there is a continued need to continue evolving RT modalities into safer and more effective treatments. The present study compares the impact of the broad beam (BB) and the spatially fractionated modality of microbeam radiation therapy (MRT) on the motility of A549 lung cancer cells. Our data corroborates previous findings that showed BB irradiation is a promoter of cell motility. For MRT, however, we observed a prevention of cellular migration. A significant reduction in NF-κB expression was observed only when A549 cells were irradiated with MRT, indicating a potential mechanism behind these findings. Finally, our data supports potential issues regarding MRT irradiation of key components of the tumor microenvironment, such as fibroblasts. Co-culturing A549 cells with MRT-irradiated MRC-5 lung fibroblasts led to increased tumor cell invasion, not observed when the fibroblasts received BB irradiation. Full article
(This article belongs to the Special Issue Cell Migration and Invasion)
12 pages, 3854 KB  
Article
Crosstalk of Tumor-Derived Extracellular Vesicles with Immune Recipient Cells and Cancer Metastasis
by Han Jie, Alicja C Gluszko and Theresa L. Whiteside
Cancers 2026, 18(2), 196; https://doi.org/10.3390/cancers18020196 - 7 Jan 2026
Abstract
Background. Contributions of tumor-derived extracellular vesicles, TEX, to tumor progression and metastasis involve their crosstalk with immune cells in the tumor microenvironment. This crosstalk results in metabolic reprogramming of immune cells from anti-tumor to pro-tumor activity. Mechanistic underpinnings of the TEX entry [...] Read more.
Background. Contributions of tumor-derived extracellular vesicles, TEX, to tumor progression and metastasis involve their crosstalk with immune cells in the tumor microenvironment. This crosstalk results in metabolic reprogramming of immune cells from anti-tumor to pro-tumor activity. Mechanistic underpinnings of the TEX entry and delivery of molecular signals responsible for metabolic reprogramming may be unique for different types of immune cells. Methods. An in vitro model of THP-1 myeloid cells co-incubated with TEX illustrates the role TEX play in polarization of macrophages to TAMs. Results. In THP-1 cells, the dominant signaling pathway of melanoma cell-derived TEX involves HSP-90/TLR2. This leads to activation of the NF-κB and MAP kinase pathways and initiates THP-1 cell polarization from M0 to M2 with strong expression of immunosuppressive PD-L1. TEX may be seen as “danger” by the myeloid cells, which utilize the pattern recognition receptors (PRR), such as PAMPs or DAMPs, for engaging the complementary ligands carried by TEX. The same melanoma TEX signaling to T cells via DAMPs induced mitochondrial stress, resulting in T-cell apoptosis. Conclusions. As the signaling receptors/ligands in TEX are determined by the tumor, it appears that the tumor equips TEX with an address recognizing specific PRRs expressed on different recipient immune cells. Thus, TEX, acting like pathogens, are equipped by the tumor to alter the context of intercellular crosstalk and impose a distinct autophagy-not-apoptosis signature in recipient THP-1 cells. The tumor might endorse TEX to promote tumor progression and metastasis by enabling them to engage the signaling system normally used by immune cells for defense against pathogens. Full article
(This article belongs to the Special Issue Exosomes in Cancer Metastasis (2nd Edition))
Show Figures

Figure 1

28 pages, 2111 KB  
Review
Integrative Sequencing and Proteogenomic Approaches to Intratumoral Heterogeneity in Cholangiocarcinoma: Implications for Precision Diagnosis and Therapy
by Sirinya Sitthirak, Arporn Wangwiwatsin, Apinya Jusakul, Nisana Namwat, Poramate Klanrit, Sittiruk Roytrakul, Hasaya Dokduang, Thitinat Duangchan, Yanisa Rattanapan, Attapol Titapun, Apiwat Jareanrat, Vasin Thanasukarn, Natcha Khuntikeo, Teh Bin Tean, Luke Boulter, Yoshinori Murakami and Watcharin Loilome
Med. Sci. 2026, 14(1), 30; https://doi.org/10.3390/medsci14010030 - 7 Jan 2026
Abstract
Cholangiocarcinoma (CCA) is a highly aggressive cancer of the biliary tract, distinguished by significant intratumoral heterogeneity (ITH), which contributes to therapy resistance and unfavorable clinical outcomes. Traditional genome profiling has revealed recurring driver changes in CCA; yet, genomic data alone fails to elucidate [...] Read more.
Cholangiocarcinoma (CCA) is a highly aggressive cancer of the biliary tract, distinguished by significant intratumoral heterogeneity (ITH), which contributes to therapy resistance and unfavorable clinical outcomes. Traditional genome profiling has revealed recurring driver changes in CCA; yet, genomic data alone fails to elucidate functional pathway activation, adaptive signaling, and the diverse treatment responses reported among tumor locations and disease subtypes. This review analyses the use of integrated sequencing technologies, proteogenomics, and phosphoproteomics to systematically characterize intratumoral heterogeneity in cholangiocarcinoma and convert molecular diversity into therapeutically applicable discoveries. We present evidence that the combination of genomic sequencing and mass spectrometry–based proteomics facilitates the direct correlation of genetic mutations with protein expression, post-translational modifications, and signaling system activity. Phosphoproteomic profiling specifically offers functional insights into kinase-driven networks that dictate tumor aggressiveness, therapeutic susceptibility, and adaptive resistance mechanisms, which cannot be anticipated only from DNA-level analysis. We propose that integrating proteogenomic and phosphoproteomic analyses into diagnostic and therapeutic assessments can enhance molecular classification, reveal subtype- and region-specific therapeutic dependencies, and guide rational combination treatment strategies, based on recent extensive proteogenomic studies and functional proteomic investigations in CCA. Pathway-level analysis of intratumoral heterogeneity provides a framework for selecting targeted medicines, predicting resistance, and informing personalized treatment strategies in CCA. The combination of sequencing, proteogenomics, and phosphoproteomics is essential for advancing precision oncology in cholangiocarcinoma. The implementation of this multi-layered analytical approach may better patient classification, refine therapy choices, and eventually improve clinical outcomes for individuals with this particular heterogeneous cancer. Full article
(This article belongs to the Section Cancer and Cancer-Related Research)
Show Figures

Figure 1

14 pages, 922 KB  
Review
Pre-Existing Immunity Shapes Cancer Immunotherapy Efficacy
by Anastasia Xagara, Filippos Koinis, Konstantinos Tsapakidis, Ioannis Samaras, Evangelia Chantzara, Konstantina Vasilieva, Alexandros Lazarou, Vassilis Georgoulias and Athanasios Kotsakis
Onco 2026, 6(1), 4; https://doi.org/10.3390/onco6010004 - 7 Jan 2026
Abstract
Immunotherapy has revolutionized the management of patients with cancer. Immune checkpoint inhibition (ICI) is a promising treatment option that targets the molecular mechanisms that cancer cells exploit to prevent immune-mediated elimination. ICI therapy can cause exceptional long-term tumor remissions, in some cases, even [...] Read more.
Immunotherapy has revolutionized the management of patients with cancer. Immune checkpoint inhibition (ICI) is a promising treatment option that targets the molecular mechanisms that cancer cells exploit to prevent immune-mediated elimination. ICI therapy can cause exceptional long-term tumor remissions, in some cases, even after treatment discontinuation. Despite its success, many patients acquire resistance or fail to respond due to immune escape mechanisms mediated by the tumor and its microenvironment. Pre-existing immunity status of individuals seems to play a fundamental role in immunotherapy response and eventually tumor progression, as it orchestrates tumor-immune interactions. Different immune cell subsets, both in the tumor microenvironment and the peripheral blood, are established mediators that contribute to immune escape in various tumor types. Based on these findings, the elucidation of the mechanisms implicated in the regulation of these immune cells has become a priority for investigators focused on improving the efficacy of ICI. This will be essential for identifying responders as well as for developing novel therapeutic modalities to improve clinical outcomes. Herein, we summarize preclinical and clinical evidence proposing a predictive role of pre-existing immunity for clinical responses to immunotherapies. Full article
(This article belongs to the Special Issue Liquid Biopsy and Peripheral Immune Status in Cancer Therapy Response)
Show Figures

Figure 1

48 pages, 1505 KB  
Review
The Role of Natural Compounds in Bladder Urothelial Carcinoma Treatment
by Hangfei Jiang, Yueyin Chen, Xi Zeng, Rui Yang, Feng Zhang, Huanling Zhang, Erxiang Zhang, Xuzhang Wu, Deye Yan and Chunping Yu
Int. J. Mol. Sci. 2026, 27(2), 596; https://doi.org/10.3390/ijms27020596 - 7 Jan 2026
Abstract
Bladder urothelial carcinoma (BUC) ranks among the most common malignant tumors of the urinary system, with alarmingly high incidence and mortality rates. Current clinical treatments face challenges such as strong chemotherapy resistance and limited response rates to immunotherapy, creating an urgent need for [...] Read more.
Bladder urothelial carcinoma (BUC) ranks among the most common malignant tumors of the urinary system, with alarmingly high incidence and mortality rates. Current clinical treatments face challenges such as strong chemotherapy resistance and limited response rates to immunotherapy, creating an urgent need for novel alternative therapies. Natural products, characterized by multi-targeted antitumor activity, low toxicity, and broad availability, have emerged as highly promising adjunctive or alternative strategies in cancer treatment. Extensive research has elucidated the antitumor activities of natural products, including inhibition of cancer cell proliferation, induction of apoptosis, and modulation of the immune microenvironment. What’s more, their bioactive components, such as terpenoids and polyphenols, can synergistically enhance therapeutic efficacy while reducing toxicity risks associated with traditional therapies. This review will examine the roles of terpenoids, phenolics, alkaloids, and other natural products in BUC treatment, to provide directions for future research. Full article
(This article belongs to the Section Bioactives and Nutraceuticals)
21 pages, 652 KB  
Review
The Role of microRNAs as Potential Biomarkers in Diffuse Large B-Cell Lymphoma
by Eirini Panteli, Epameinondas Koumpis, Vasileios Georgoulis, Georgios Petros Barakos, Evangelos Kolettas, Panagiotis Kanavaros, Alexandra Papoudou-Bai and Eleftheria Hatzimichael
Non-Coding RNA 2026, 12(1), 2; https://doi.org/10.3390/ncrna12010002 - 7 Jan 2026
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common and clinically aggressive subtype of non-Hodgkin lymphoma (NHL). While novel therapies such as rituximab and polatuzumab vedotin have led to improved outcomes, approximately 35% of patients eventually develop relapsed or refractory disease. MicroRNAs (miRNAs), [...] Read more.
Diffuse large B-cell lymphoma (DLBCL) is the most common and clinically aggressive subtype of non-Hodgkin lymphoma (NHL). While novel therapies such as rituximab and polatuzumab vedotin have led to improved outcomes, approximately 35% of patients eventually develop relapsed or refractory disease. MicroRNAs (miRNAs), a class of endogenous single-stranded RNAs approximately 22 nucleotides in length, play a pivotal role in the regulation of gene expression at the post-transcriptional level through interactions with complementary target RNAs and contribute significantly to the development, progression, and treatment response of DLBCL. Oncogenic miRNAs, such as miR-155, miR-21, and the miR-17–92 cluster, promote proliferation, survival, immune evasion, and therapy resistance by modulating pathways including PI3K/AKT, NF-κB, and MYC. Conversely, tumor-suppressive miRNAs such as miR-34a, miR-144, miR-181a, and miR-124-3p inhibit oncogene activity and enhance apoptosis, with their loss often associated with adverse outcomes. Among these, miR-155 and miR-21 are particularly well studied, playing central roles in both tumor progression and remodeling of the tumor microenvironment. This review summarizes current evidence on the biological and clinical relevance of miRNAs in DLBCL, emphasizing their diagnostic and prognostic potential. Full article
Show Figures

Figure 1

20 pages, 1618 KB  
Review
Cholesterol Metabolism: An Ally in the Development and Progression of Cervical Cancer
by Imelda Martínez-Ramírez, J. Omar Muñoz-Bello, Adriana Contreras-Paredes, Elías Parra-Hernández, Adela Carrillo-García and Marcela Lizano
Int. J. Mol. Sci. 2026, 27(2), 591; https://doi.org/10.3390/ijms27020591 - 6 Jan 2026
Abstract
Despite screening programs and vaccination campaigns, cervical cancer (CC) remains a health problem worldwide. The involvement of the E6 and E7 oncoproteins of Human Papillomavirus (HPV) is crucial for the development and progression of this type of cancer. Metabolic reprogramming by cancer cells [...] Read more.
Despite screening programs and vaccination campaigns, cervical cancer (CC) remains a health problem worldwide. The involvement of the E6 and E7 oncoproteins of Human Papillomavirus (HPV) is crucial for the development and progression of this type of cancer. Metabolic reprogramming by cancer cells has gained relevance in the last decade due to its ability to promote cell growth, survival, invasion, metastasis, and resistance to therapy. In this review, we focus on alterations in cholesterol metabolism that significantly influence the development and progression of CC, as well as the clinical outcome of patients. Furthermore, evidence from comprehensive omics studies suggesting that E6 and E7 are involved in the exacerbation of elements related to cholesterol metabolism is analyzed. Preclinical and clinical studies are also discussed that demonstrate that cholesterol metabolism is a potential therapeutic target, highlighting its impact on reducing tumor growth, altering the tumor microenvironment, and improving antitumor immunity. Full article
(This article belongs to the Special Issue Molecular Insights and Treatments for Gynecological Cancers)
Show Figures

Figure 1

21 pages, 4482 KB  
Article
X-Ray-Induced Alterations in In Vitro Blood–Brain Barrier Models: A Comparative Analysis
by Roberta Moisa (Stoica), Stela Rodica Lucia Pătrașcu, Călin Mircea Rusu, Mihail Răzvan Ioan, Mihai Radu and Beatrice Mihaela Radu
Appl. Sci. 2026, 16(2), 587; https://doi.org/10.3390/app16020587 - 6 Jan 2026
Abstract
Ionizing radiation remains the primary approach for treating brain cancer and is frequently used in combination with chemotherapy. However, when it comes to gliomas, the effective delivery of therapeutic agents is hindered by the limited permeability of the blood–brain barrier (BBB). Consequently, selecting [...] Read more.
Ionizing radiation remains the primary approach for treating brain cancer and is frequently used in combination with chemotherapy. However, when it comes to gliomas, the effective delivery of therapeutic agents is hindered by the limited permeability of the blood–brain barrier (BBB). Consequently, selecting the most suitable and least harmful type of ionizing radiation is essential, given its potential side effects on healthy cells within the tumor microenvironment. In this study, we explored the impact of X-ray exposure on two in vitro BBB endothelial cell models—murine and human. Post-irradiation, we evaluated cell viability, clonogenic capacity, cell cycle progression, reactive oxygen species (ROS) levels, formation of micronuclei and γ-H2AX foci, as well as alterations in cytoskeletal organization, cell migration, and intracellular calcium dynamics. The results demonstrate notable differences between the two endothelial cell lines, suggesting the human cell line is more sensitive to X-rays. In conclusion, our study provides valuable insights into the brain microvascular endothelial cells' response to radiation, laying the groundwork for strategies to protect healthy brain tissue. Full article
(This article belongs to the Special Issue Radiation Physics: Advances in DNA and Cellular Technologies)
54 pages, 3566 KB  
Review
Implementation of Natural Products and Derivatives in Acute Myeloid Leukemia Management: Current Treatments, Clinical Trials and Future Directions
by Faten Merhi, Daniel Dauzonne and Brigitte Bauvois
Cancers 2026, 18(2), 185; https://doi.org/10.3390/cancers18020185 - 6 Jan 2026
Abstract
Bioactive natural products (NPs) may play a critical role in cancer progression by targeting nucleic acids and a wide array of proteins, including enzymes. Furthermore, a large number of derivatives (NPDs), including semi-synthetic products and pharmacophores from NPs, have been developed to enhance [...] Read more.
Bioactive natural products (NPs) may play a critical role in cancer progression by targeting nucleic acids and a wide array of proteins, including enzymes. Furthermore, a large number of derivatives (NPDs), including semi-synthetic products and pharmacophores from NPs, have been developed to enhance the solubility and stability of NPs. Acute myeloid leukemia (AML) is a poor-prognosis hematologic malignancy characterized by the clonal accumulation in the blood and bone marrow of myeloid progenitors with high proliferative capacity, survival and propagation abilities. A number of potential pathways and targets have been identified for development in AML, and include, but are not limited to, Fms-like tyrosine kinase 3 (FLT3) and isocitrate dehydrogenases resulting from genetic mutations, BCL2 family members, various signaling kinases and histone deacetylases, as well as tumor-associated antigens (such as CD13, CD33, P-gp). By targeting nucleic acids, FLT3 or CD33, several FDA-approved NPs and NPDs (i.e., cytarabine, anthracyclines, midostaurin, melphalan and calicheamicin linked to anti-CD33) are the major agents of upfront treatment of AML. However, the effective treatment of the disease remains challenging, in part due to the heterogeneity of the disease but also to the involvement of the bone marrow microenvironment and the immune system in favoring leukemic stem cell persistence. This review summarizes the current state of the art, and provides a summary of selected NPs/NPDs which are either entering or have been investigated in preclinical and clinical trials, alone or in combination with current chemotherapy. With multifaceted actions, these biomolecules may target all hallmarks of AML, including multidrug resistance and deregulated metabolism. Full article
(This article belongs to the Special Issue Study on Acute Myeloid Leukemia)
Show Figures

Figure 1

19 pages, 1075 KB  
Review
Circadian Clock Genes in Colorectal Cancer: From Molecular Mechanisms to Chronotherapeutic Applications
by Haoran Wang, Jieru Zhou, Suya Pang, Yiqing Mei, Gangping Li, Yu Jin and Rong Lin
Biomedicines 2026, 14(1), 110; https://doi.org/10.3390/biomedicines14010110 - 6 Jan 2026
Viewed by 20
Abstract
Colorectal cancer (CRC) is a life-threatening malignancy, but our understanding of its pathogenic mechanisms remains incomplete—posing a major constraint on the development of effective therapeutic strategies. The transcription-translation feedback loop of clock genes (e.g., BMAL1, CLOCK, PER1/2/3, and CRY1/ [...] Read more.
Colorectal cancer (CRC) is a life-threatening malignancy, but our understanding of its pathogenic mechanisms remains incomplete—posing a major constraint on the development of effective therapeutic strategies. The transcription-translation feedback loop of clock genes (e.g., BMAL1, CLOCK, PER1/2/3, and CRY1/2) provides a promising novel avenue for deciphering the initiation and progression of CRC. Mounting evidence indicates that core circadian clock genes play pivotal roles in CRC oncogenesis by orchestrating the regulation of the cell cycle, epithelial–mesenchymal transition (EMT), metabolic reprogramming, and the tumor microenvironment. This review systematically summarizes the expression patterns and mechanistic roles of core clock genes in CRC, while elucidating their molecular underpinnings in tumor progression via key signaling cascades (e.g., Wnt/β-catenin and c-Myc/p21 pathways). We emphasize the associations between circadian disruption and CRC—including diagnostic markers, prognostic assessment, and chemosensitivity—and provide an in-depth discussion of chronotherapeutic strategies and their translational potential. Finally, we identify unaddressed scientific questions and propose future research directions to facilitate the development of novel targeted therapies for CRC. Full article
(This article belongs to the Special Issue Advancements in the Treatment of Colorectal Cancer)
Show Figures

Graphical abstract

Back to TopTop