Current Cancer Drug Targets - Online First
Description text for Online First listing goes here...
1 - 20 of 71 results
-
-
Overexpression of HNF4G can Increase the Resistance of Ovarian Cancer Cells to Olaparib
Authors: Ying Dong, Yue Deng, Huilin Yang, Youfang Hou, Qin Zhang and Lihua YangAvailable online: 17 October 2025More LessIntroductionOvarian cancer (OV) is one of the most malignant gynecological cancers. Poly(ADP-ribose) polymerase inhibitors (PARPi) represent the first-line maintenance therapy, effectively prolonging patient survival; however, the development of PARPi resistance poses a significant challenge for OV maintenance therapy. Previous studies have indicated that HNF4G functions as an oncogene in various tumors, but its role in OV development and Olaparib resistance remains unexplored.
MethodsWe established an Olaparib-resistant OV cell line, SKOV3-PARPi, from the parental SKOV3 cell line. The impact of HNF4G on SKOV3 cell resistance to Olaparib was investigated using qRT-PCR, CCK-8 assay, Transwell assay, colony formation assay, scratch assay, Western blot, flow cytometry, as well as a nude mouse xenograft tumor model and immunohistochemistry. The function of HNF4G in SKOV3-Olaparib resistant cells was elucidated and subsequently validated through the animal tumor model.
ResultsProlonged Olaparib exposure induced acquired resistance in SKOV3 cells. Compared to parental OV cells, HNF4G expression was upregulated in Olaparib-resistant cells. Overexpression of HNF4G enhanced Olaparib resistance in OV cells, whereas HNF4G knockdown diminished it. Furthermore, increased protein levels of components within the PI3K-AKT signaling pathway were observed in Olaparib-resistant cells. Knocking down HNF4G expression in resistant cells significantly slowed tumor growth under Olaparib treatment. Changes in the protein levels of HNF4G and PI3K-AKT pathway components in the in vivo xenograft tumor tissues were consistent with the cellular observations.
ConclusionOverexpression of HNF4G plays a crucial role in conferring Olaparib resistance in OV by activating the PI3K-AKT signaling pathway. HNF4G may serve as a potential therapeutic target for patients with Olaparib-resistant OV.
-
-
-
Gut Microbiota: An Innovative Traditional Chinese Medicine Perspective on Breast Cancer Therapy
Authors: Haining Ding, Yian Chen, Qinghong Yu, Hailong Li and Xiufei GaoAvailable online: 08 October 2025More LessBreast cancer (BC) represents a complex malignancy shaped by both genetic predisposition and environmental influences, with growing evidence implicating the gut microenvironment in its pathogenesis. While the therapeutic potential of gut-targeted interventions has gained attention, the precise molecular mechanisms remain poorly characterized. Traditional Chinese medicine (TCM) has emerged as a valuable therapeutic approach due to its widespread availability and demonstrated clinical efficacy, particularly through its capacity to modulate gut homeostasis and exert systemic effects across multiple disease states, including breast cancer. Specific TCM formulations, including CCM, CMM, and MBC, have shown significant potential to reshape gut microbial composition, influence microbial metabolite production, restore immune homeostasis, enhance short-chain fatty acid biosynthesis, regulate estrogen metabolism, and induce beneficial epigenetic modifications, thereby offering a multifaceted therapeutic strategy against breast cancer. This review systematically examines the pharmacological mechanisms, molecular targets, and clinical implications of TCM-based interventions in breast cancer management, highlighting their potential to open new avenues in oncological therapeutics.
-
-
-
Zinc-Facilitated Enzyme Disruption: Thiazolidinediones as Potent Carbonic Anhydrase Inhibitors in Hypoxic Cancer Microenvironments
Authors: Gaurav Ranjan, Shakti Prasad Pattanayak and Priyashree SunitaAvailable online: 08 October 2025More LessIntroductionThiazolidinedione (TZD) derivatives have gained significant attention as anti-cancer agents due to their diverse biological activities. The objective of the research was to investigate the potential of thiazolidine derivatives as inhibitors of Carbonic anhydrase XII, an isoform overexpressed in hypoxic tumor environments, to explore their application as anticancer agents targeting breast cancer.
MethodsThe study employed a computational approach to evaluate thiazolidine derivatives as potential CA XII inhibitors. Acute toxicity and safety were evaluated using ProTox 3.0. Molecular docking was conducted to study interactions with the zinc-bound active site of CA XII. Molecular dynamics simulations were performed to validate the stability of the ligand-enzyme complex over 250 ns.
ResultsTZD derivatives demonstrated favorable physicochemical properties, high gastrointestinal absorption, and low toxicity risks. Molecular docking studies showed strong binding affinities with key hydrogen bonding and zinc coordination at the CA XII active site. Toxicity predictions indicated that most compounds had acceptable safety margins, reinforcing their potential as safe and effective CA XII inhibitors.
DiscussionThe findings suggest that thiazolidine scaffolds could serve as promising small-molecule inhibitors of CA XII by targeting its zinc-bound catalytic site, a mechanism consistent with previously reported CA inhibitor pharmacology.
ConclusionThe study demonstrates that TZD derivatives possess promising characteristics as CA XII inhibitors, with favorable physicochemical properties, strong binding affinity, stable ligand-protein interactions, and acceptable safety profiles. These findings highlight their potential for further in vitro and in vivo validation, supporting the continued exploration of thiazolidinedione scaffolds in the development of targeted anticancer therapies.
-
-
-
Multi-omics Approaches to CCAAT/Enhancer-Binding Protein Beta in Oral Squamous Cell Carcinoma: Crosstalk Between Tumor Cells andTumor-Associated Macrophages Driving Disease Progression
Authors: Min Li, Jilun Liu, Wenjuan Zhang, Ruonan Sun, Wenjing Wang, Xin Liu, Linyu Jin and Yongle QiuAvailable online: 18 September 2025More LessBackgroundCCAAT/Enhancer-Binding Protein Beta (CEBPB) is an important transcription factor that regulates tumor progression. However, the mechanism by which CEBPB regulates the progression of Oral Squamous Cell Carcinoma (OSCC) remains incompletely understood. Tumor progression depends on complex intercellular interactions within the tumor microenvironment. The purpose of this study was to investigate the role and epigenetic regulatory mechanisms of CEBPB in interactions between OSCC cells and tumor-infiltrating immune cells.
MethodsBulk RNA-seq, ChIP-seq, and scRNA-seq data were obtained from The Cancer Genome Atlas (TCGA) database and the Gene Expression Omnibus (GEO) database. The HOMER algorithm was employed to identify enhancers and predict the CEBPB-binding motif. Cell cluster analysis, functional enrichment, and intercellular interaction analysis were performed using the “Seurat” R package. H3K27ac enrichment at GAS6 enhancers was validated by ChIP-qPCR. Metastatic OSCC cells with CEBPB knockdown or GAS6 overexpression were established and co-cultured with THP-1 cells. IL-10 and IL-6 secretion from co-cultured THP-1 cells was detected via ELISA. Chemotaxis of OSCC cells toward THP-1 cells was assessed through a Transwell assay.
ResultsCEBPB was upregulated in OSCC and correlated with poor prognosis. By integrating H3K27ac ChIP-seq and bulk RNA-seq data, 131 CEBPB-regulated enhancer-controlled genes were identified in lymph node metastatic OSCC cells. scRNA-seq analysis revealed eight major cell clusters in primary foci and lymph node metastases, including T/NK cells, malignant epithelial cells, B/plasma cells, macrophages, fibroblasts, dendritic cells, endothelial cells, and mast cells, with the malignant epithelial cells stratified into distinct sub-clusters. CEBPB expression was elevated in malignant epithelial cells of lymph node metastases compared to primary foci. Furthermore, 15 pairs of enhanced ligand-receptor interactions were identified in lymph node metastases relative to primary foci. GAS6 was a CEBPB-regulated enhancer-controlled gene, primarily mediating interactions between malignant cells and macrophages. CEBPB knockdown in metastatic OSCC cells significantly impaired their chemotaxis toward co-cultured THP-1 cells, and downregulated IL-10/IL-6 secretion and CD206 expression in co-cultured THP-1 cells. Conversely, GAS6 overexpression reversed these inhibitory effects.
ConclusionCEBPB activated GAS6 transcription in metastatic OSCC cells. The CEBPB/GAS6 axis in metastatic OSCC cells enhanced their chemotaxis toward macrophages and promoted the M2 polarization of macrophages, thereby facilitating the establishment of an immunosuppressive microenvironment.
-
-
-
Driver Genes and Genomic Instability Predict the Incidence and Outcome of Brain Metastases
Authors: Hainan Yang, Weiping Hong, Jie Ding, Weifang Yuan, Hui Ye, Tao Lin, Qingjun Hu, Xin Jin, Lei Wen, Da Liu and Ming LeiAvailable online: 03 September 2025More LessIntroductionThe incidence of brain metastases in patients diagnosed with advanced lung cancer is high, drawing significant attention to the risk factors associated with this progression.
MethodsA total of 252 advanced non-small cell lung cancer (NSCLC) patients with brain metastases were enrolled in this study between July 2018 and December 2023 from our hospital. Additionally, driver genes, including EGFR, ALK, ROS1, KRAS, and RET, were documented. Next-generation targeted sequencing of a 168-gene panel was conducted on all collected samples to explore the association between tumor genomic complexity and risk factors for NSCLC with brain metastases.
ResultsAmong 252 lung cancer patients with brain metastases enrolled in this research, the most prevalent driver gene was EGFR, accounting for 39.29% (99 patients). Other driver gene mutations, such as KRAS, ALK, ROS1, and RET, accounted for 3.57%, 7.14%, 2.78%, and 0.4%, respectively. Kaplan-Meier analysis showed that patients with EGFR mutations had a more favorable overall survival (OS) compared to those without the mutation (P < 0.0001). Additionally, patients with ALK fusions had longer survival times compared to those with wild-type genes (P = 0.0021). In this study, patients were divided into two groups based on the presence or absence of copy-number alterations. Further survival analysis revealed that patients with copy-number alterations experienced significantly shorter overall survival compared to the control group (P = 0.041).
DiscussionThis study underscores the crucial role of driver mutations and genomic instability in advanced NSCLC with brain metastases, where EGFR and ALK alterations are linked to better survival. In contrast, high genomic complexity is associated with worse outcomes.
ConclusionDriver gene mutations are present in more than half of the patients with central nervous system (CNS) failure. Genomic instability, characterized by the number of co-occurring mutated genes and copy-number alterations, is a risk factor associated with shorter survival time.
-
-
-
High Glucose and Glucose-derived Intermediates are Linked to Lung Cancer Aggressiveness
Authors: Himani Joshi, Raiyan Satti and M. Saeed SheikhAvailable online: 29 August 2025More Less
-
-
-
Comparing Ovarian Clear Cell Carcinoma and High-Grade Serous Carcinoma Based on the SEER Database and Analyzing the Significantly Mutated Genes
Authors: Xuzhi Liang, Ying Yang, Shiyu Zhang, Haijing He, Yuqi Wen and Jiangtao FanAvailable online: 28 August 2025More LessIntroductionOvarian clear cell carcinoma (OCCC) accounts for about 5% of all epithelial ovarian cancers. Currently, its treatment mainly refers to high-grade serous carcinoma (HGSC). This study aimed to explore differences in clinical characteristics between OCCC and HGSC and studied the reasons for the differences.
MethodsThe data of OCCC and HGSC cases were obtained from the SEER database. Univariate and multivariate Cox regression analyses were used to explore the prognostic factors. Next, whole exome sequencing (WES) was performed on 15 clinically selected OCCC cases and 16 HGSC cases to identify significantly mutated genes (SMGs). Further analysis included calculating tumor mutation burden (TMB) and predicting potential target drugs based on the identified mutations.
Results3493 OCCC and 10266 HGSC patients from the SEER database were included in the study. Survival analysis showed that the overall survival (OS) of stage I-II OCCC was better than that of stage I-II HGSC, while the OS of stage III-IV OCCC was worse than that of stage III-IV HGSC. Further subgroup analysis showed that for the OCCC group, age ≥ 60 years, bilateral tumor distribution, tumor size ≥ 87mm, and stage III-IV were independent risk factors for OS. For HGSC patients, tumor size ≥ 87mm was an independent protective factor for OS. WES results suggested that among the top 20 SMGs of OCCC in stage III-IV patients, DNAH2, LAMA5, MUC19, NOTCH1, PCLO, SYNE2, TACC2, and ZNF469 were 8 specific SMGs that distinguish III-IV OCCC from III-IV HGSC. In addition, the stage I-II OCCC group had the highest TMB, and the lowest was the stage III-IV OCCC.
DiscussionOur findings challenge the conventional uniform therapeutic approach for ovarian carcinomas by revealing stage-dependent SMGs between OCCC and HGSC. However, limitations such as the retrospective SEER analysis, small WES cohort, and population-specific driver gene variations require cautious interpretation of the findings.
ConclusionThe independent prognostic factors identified in this study provide a theoretical basis for individualized prognosis judgment in OCCC and HGSC. The SMGs and TMB levels may serve as valuable indicators for prognosis and evaluating targeted therapy or immunotherapy efficacy. Druggable genes such as NOTCH1 and RYR3 offer promising therapeutic targets, while stage-specific pathway enrichments reveal potential intervention strategies. Further validation in larger cohorts is needed to confirm these findings. Our study advances the understanding of molecular heterogeneity in ovarian cancer and lays the groundwork for personalized treatment strategies, ultimately improving patient outcomes.
-
-
-
SOX9 Promotes Breast Cancer Progression via the EGFR/STAT3 Signaling Axis
Authors: Chunrui Zhang, Na Li, Fei Xue and Tipeng ZhangAvailable online: 22 August 2025More LessIntroductionSex-determining region Y-box 9 (SOX9) is a transcription factor frequently overexpressed in breast cancer, playing a critical role in tumor initiation, progression, and therapeutic resistance. While its oncogenic potential is recognized, the underlying molecular mechanisms remain incompletely elucidated. This study aimed to investigate the functional role of SOX9 in breast cancer, specifically focusing on its interaction with the EGFR/STAT3 signaling pathway.
MethodsThe study integrated bioinformatics analyses with functional assays in breast cancer cell lines to determine the effects of SOX9 modulation on cell proliferation, migration, and invasion, and to elucidate its connection with the EGFR/STAT3 signaling axis.
ResultsOur findings demonstrate that SOX9 promotes breast cancer cell proliferation, migration, and invasion. Mechanistically, this occurs through the activation of the EGFR/STAT3 signaling axis. Furthermore, targeting SOX9 effectively attenuated these oncogenic phenotypes in vitro.
DiscussionThe elucidation of SOX9’s role in activating the EGFR/STAT3 pathway significantly advances our understanding of its oncogenic mechanisms in breast cancer. These findings are consistent with existing literature on SOX9’s pro-tumorigenic impact and the established role of EGFR/STAT3 signaling in cancer progression, highlighting a crucial regulatory link. This newly identified SOX9-EGFR/STAT3 axis not only reinforces SOX9’s prognostic value but also strongly supports its exploration as a novel therapeutic target.
ConclusionThese findings identify SOX9 as a key regulator of the EGFR/STAT3 signaling pathway in breast cancer. This highlights the potential of SOX9 as both a prognostic biomarker and a promising target for drug therapy in breast cancer.
-
-
-
Single-Cell Transcriptomics: Technical Advances, Applications and Challenges in Cancer Drug Discovery
Authors: Yueying Yang, Lingyu Meng, Teng Zhang and Jianjun TanAvailable online: 30 July 2025More LessWith advancements in technology, single-cell RNA sequencing has emerged as a powerful tool in cancer drug discovery. This technique enables the construction of gene expression profiles at the individual cell level, offering detailed insights into cellular heterogeneity and molecular pathways involved in tumor development. It enables researchers to gain a deeper understanding of tumor heterogeneity. Researchers can study cell subpopulations and gene expression patterns. This understanding helps in identifying potential drug targets. Additionally, it aids in predicting therapeutic responses. This high-resolution gene expression analysis provides a new perspective and opportunity for cancer drug development, which is expected to accelerate the discovery and development process of new anti-cancer drugs. This article provides a comprehensive overview of the basic processes and developmental trajectory of single-cell RNA sequencing technology, with a particular emphasis on its applications in various aspects of cancer drug discovery. It also addresses the challenges faced by single-cell RNA sequencing and potential future directions. This review aims to enhance readers’ understanding of single-cell sequencing, inspire new ideas for oncology drug development, and support the translation of clinical research into practice, ultimately enabling physicians to design more precise and personalized treatment strategies.
-
-
-
To Combat Abnormal Cell Signaling Mediated Gastrointestinal Cancer by Therapeutic Modulation of Gut Microbiota Utilizing Prebiotics, Postbiotics, and Synbiotics
Authors: Devasmita Sen, Sristi Datta, Srija Biswas, Asmita Samadder and Sisir NandiAvailable online: 23 July 2025More LessBackgroundGastrointestinal (GI) cancer, a multifactorial disease, encompasses a group of malignancies that affect the gastrointestinal system. Being the second leading contributor to cancer-related deaths, GI cancer has become the burning issue of human health. Despite advances in treatment, the diverse nature of GI cancer indicates that a one-size-fits-all solution is not applicable.
IntroductionThe gut microbiome can be therapeutically modulated by utilizing prebiotics, postbiotics, and synbiotics. Fermentation of prebiotics produces postbiotic compounds. Together the prebiotics and probiotics combination can be used as synbiotics which will be more beneficial.
MethodsPubMed and Google scholar search engine tools have been utilized to access references about the idea of this review to demonstrate the therapeutic modulation of microbiota, residing in the gut, which utilizes postbiotics, prebiotics and synbiotics for combating GI cancer.
ResultsExploration of prebiotics, postbiotics, and synbiotic compounds has given us detailed information about their contribution to combating GI cancer.
ConclusionIntake of a combination of prebiotic, postbiotics and synbiotics can inhibit the growth of cancer cells, and activate protective and stress-resistant mechanisms in healthy cellswhich couldbe more beneficial than the administration of prebiotics or postbiotics or synbiotics alone in diminishing the risk of GI cancer.
-
-
-
Migrasomes: A Novel Target for Cancer Treatment
Authors: Neha Sharma and Anurag ChaudharyAvailable online: 22 July 2025More LessProtrusion and adhesion occur at the foremost point of cells during cell migration, while contraction and detachment occur at the rear of the cells. The combined action of cytoskeletal dynamics, vesicular trafficking, and signaling networks initiates this multi-step process. The development of a novel exosome-like organelle called migrasomes, which may play roles in intercellular signaling, and which originate from retraction fibers at the back of migrating cells. Migrasomes are a particular kind of extracellular vesicle that is placed by a special mechanism and left behind by migrating cells. The proteins called integrins, which connect cells to the extracellular matrix (ECM), regulate the mobilization of migrasome. The function of migrasomes is to preserve cellular homeostasis and communication between cells. By observing this literature, we attempted to ascertain the potential role that migrasomes will play in the future in illnesses involving migrating cells, like immune system problems, tumor metastasis, and other disorders.
-
-
-
The Role of Lactate in Cancer Immunotherapy: Mechanisms and Applications
Authors: Yunhui Fan, Haoyue Jia and Wanguang ZhangAvailable online: 17 July 2025More LessIn recent years, immunotherapy has demonstrated significant clinical effectiveness. However, challenges such as low response rates, severe treatment-related side effects, and acquired immune tolerance persist in tumor immunotherapy. Metabolic dysregulation is acknowledged as a principal factor in tumor growth, with aerobic glycolysis, or the Warburg effect, being a defining characteristic of numerous cancers. The enhanced uptake of glucose and glycolysis provides the necessary intermediates for anabolic reactions, which are essential for the proliferation of cancer cells, while simultaneously supplying sufficient energy. However, the concomitant increase in lactate production contributes to immunosuppression within the tumor microenvironment. Tumor cells exploit lactate anabolism, lactate shuttling, and lysine lactylation modifications, which significantly diminish the efficacy of immunotherapy. The treatment targeting lactate anabolism or lactate transport proteins may prove an effective strategy for enhancing the effectiveness of cancer immunotherapy. This review provides a comprehensive overview of the role of lactate in anti-tumor immunotherapy, with the objective of deepening the understanding of the importance of lactate monitoring in cancer treatment. By elucidating these mechanisms, we aim to suggest innovative avenues for clinical cancer management, potentially improving therapeutic outcomes and overcoming the existing limitations of immunotherapy.
-
-
-
CD133+-Derived Exosomes Carrying EIF3B Mediate Cell Metastasis and Stemness in Colorectal Cancer
Authors: Xiangwei Liao, Xiaodong Han, Yu Wang, Jun Yan and Zhenqian WuAvailable online: 11 July 2025More LessBackgroundColorectal cancer (CRC) is among the most widespread malignancies worldwide and is a leading cause for cancer mortality. The interstitial interaction between cancer and stem cells is important during cancer cell metastasis.
ObjectiveIn this study, we aimed to elucidate the regulatory role and the underlying mechanisms controlling the activity of exosomes derived from cancer stem cells (CSCs).
MethodsOur group isolated exosomes from CSCs and non-CSCs to examine their regulatory mechanisms using Transwell migration, Cell Counting Kit-8 (CCK-8), and 5-ethynyl-2′-deoxyuridine (EdU) assays.
ResultsThe role of Eukaryotic Translation Initiation Factor 3 Subunit B (EIF3B) in CRC was examined using an in vivo tumorigenesis mouse model. It was found that treatment with exosomes isolated from CD133+ cells (CD133+Exos) promoted the proliferation and migration of SW480 cells. The downregulation of EIF3B reduced the proliferation and migration-promoting effects of CD133+ Exos on SW480 cells. Furthermore, CD133+ Exos treatment promoted the tumorigenesis of SW480 cells.
ConclusionOur findings demonstrate that CSC-derived exosomes transport EIF3B into CRC cells to initiate epithelial-to-mesenchymal transition (EMT) and promote metastasis.
-
-
-
Cancer Vaccines: Mechanisms, Clinical Applications, Challenges, and Future Directions in Precision Medicine
Available online: 11 July 2025More LessCancer poses a major health burden worldwide, necessitating the development of novel therapeutic approaches. Personalized cancer vaccines represent a promising form of immunotherapy that enhances the ability of the immune system to recognize and destroy tumor cells through tumor-associated and cancer-specific antigens. This review categorizes cancer vaccines into preventive, therapeutic, and personalized vaccines, discussing their mechanisms, clinical applications, and current FDA-approved examples, such as Sipuleucel-T and HPV vaccines. We highlight the recent advances in RNA-based vaccines, viral vectors, and nanotechnology, along with the synergistic role of cancer vaccines and immune checkpoint inhibitors in improving therapeutic efficacy. Overcoming ethical, regulatory, and technological barriers through global collaboration is essential for maximizing vaccine efficacy and enhancing patient outcomes. This review highlights the pivotal role of personalized vaccines in advancing precision medicine and reshaping cancer treatment paradigms.
-
-
-
Identification of AR-targeted Active Compounds from Euphorbia humifusa Willd for the Treatment of Prostate Cancer
Available online: 10 July 2025More LessIntroductionEuphorbia humifusa Willd (EH) is a traditional medicinal herb in China. However, the anti-prostate cancer active compounds of EH and their molecular mechanisms have yet to be elucidated.
MethodsThe peaks of EH water extract in the fingerprinting were analysed using liquid chromatography coupled to quadrupole time of flight mass spectrometry. The cell viability of 22RV1 cells was determined via MTT. The active compounds and potential targets were screened in silico. The prostate cancer-associated targets were collected from the GeneCards database. The herb-compound-target-disease (H-C-T-D) and PPI networks were constructed to predict key targets. The molecular docking analysis of the active compounds with key targets was conducted using Autodock Vina 1.1.2. Western blot analysis was performed to evaluate the protein expression.
ResultsLC-MS results demonstrated that EH water extract is a rich source of phenolics and flavonoids. EH water extract inhibited the viability of 22RV1 cells in a time-and dose-dependent manner. Moreover, the in silico screening results identified 17 active compounds from EH with 518 prostate cancer-related key genes. Moreover, an H-C-T-D network analysis combined with the PPI network results effectively identified seven chemical compounds, oestrogen receptor 1, and androgen receptor (AR) to be highly related to prostate cancer. Furthermore, molecular docking results showed that 4′,5-dihydroxyflavone, ensaculin, luteolin, hypolaetin, quercetin, and kaempferol had a strong binding affinity with AR. Finally, Western blot results demonstrated that EH water extract, quercetin, kaempferol, and luteolin significantly down-regulated the AR protein expression in 22RV1 cells.
ConclusionThese results suggest that EH may provide a new promising therapeutic for prostate cancer treatment.
-
-
-
Analysis of Single-Cell RNA-Seq Data to Investigate Tumor Cell Heterogeneity in Uroepithelial Bladder Cancer and Predict Immunotherapy Response
Authors: Lu Zhang, Yu Wang and Jianjun TanAvailable online: 08 July 2025More LessBackgroundNumerous studies have suggested a close association between cancer stem cells (CSCs) and the tumor microenvironment (TME), suggesting that cancer stemness might also contribute to ICI resistance. However, the interplay between these physiological processes in urothelial bladder cancer (UBC) remains unclear.
MethodsA meta-analysis was performed using the UBC Single-cell RNA sequencing (scRNA-seq) dataset, and tumor stemness gene sets (Ste.genes) were obtained. The relationship between Ste.genes and ICI response, as well as response to drug therapy, was investigated using Tumour Immune Dysfunction and Exclusion (TIDE) and drug sensitivity analyses. Machine learning based on Ste.genes was also used to predict ICI response.
ResultsA hypoxia-related tumor subgroup associated with angiogenesis and tumor metastasis was identified, and prognostic models were constructed based on hypoxic tumor subgroups. It was also found that the Ste.genes score was associated with cellular immunity, tumor immunotherapy response, and drug sensitivity. Multiple machine learning models were used to predict ICI response based on Ste.genes, and the AUC was greater than 0.7, indicating that Ste.genes can predict ICI response effectively.
ConclusionIn this study, the analysis of UBC scRNA-seq data provided further insight into the role of hypoxic tumor subpopulations in tumor development in UBC, and a prognostic model was constructed. Additionally, an association was found between cell stemness and resistance to immunotherapy as well as drug sensitivity in UBC. Ste.genes were extracted and utilized to predict the ICI response.
-
-
-
Nucleolin as a Crucial Player in Head and Neck Cancer: Diagnostic and Therapeutic Perspectives
Available online: 02 July 2025More LessHead and Neck Cancer (HNC) encompasses a diverse group of malignancies arising in the oral cavity, pharynx, larynx, and related structures. It represents a significant global health bur- den due to its high incidence, aggressive progression, and strong associations with environmental and viral risk factors like tobacco use and HPV infection. HNC, particularly squamous cell carcinoma, ranks as the seventh most common cancer worldwide. Despite the established role of these risk factors, the molecular mechanisms driving disease progression remain underexplored, especially in the context of specific biomarkers like Nucleolin (NCL). Nucleolin, a multifunctional protein, is pivotal in cancer progression, regulating cell proliferation, angiogenesis, and apoptosis. Data from The Cancer Genome Atlas (TCGA) reveal significant overexpression of NCL in HNC, particularly in advanced stages, correlating with poor prognosis and reduced patient survival. These findings highlight its potential as a diagnostic and therapeutic target. This review provides a fresh perspective on the underexplored potential of NCL as a therapeutic target in HPV-induced HNC and oral cancer. Emerging approaches, such as the AS1411 aptamer and F3 peptide, offer promising avenues for targeting NCL, paving the way for more effective, personalized cancer therapies.
-
-
-
CircRNAs Regulate Senescence-Associated miRNAs in Gastric Cancer
Authors: Shiyu Chen, Xiaoyan Yang, Xiaoyong Lei and Huifang TangAvailable online: 01 July 2025More LessGastric cancer is closely associated with the aging process, with its incidence and mortality rates significantly increasing with age, peaking around 85 years. Despite advancements in treatment modalities, current diagnostic and therapeutic approaches remain insufficient, resulting in persistently low five-year survival rates among patients. The expanding global population and the intensifying aging process are anticipated to exacerbate the global burden of gastric cancer further, underscoring the urgency of exploring novel therapeutic strategies. A complex relationship exists between gastric cancer and cellular senescence, although the precise mechanisms remain incompletely understood. Cellular senescence is prevalent in gastric cancer treatment, typically serving as a natural anti-tumor barrier by inhibiting the uncontrolled proliferation and malignant transformation of cancer cells. However, prolonged cellular senescence may trigger the secretion of pro-inflammatory factors, thereby promoting tumorigenesis and progression. A systematic analysis of existing research data has revealed significant intersections between therapeutic targets for gastric cancer and senescence-associated signaling pathways, suggesting that modulating these critical nodes may constitute a pivotal mechanism for exploring novel therapeutic strategies bridging gastric cancer treatment and senescence. Circular RNAs (circRNAs) have garnered considerable attention with the advancement of bioinformatics and high-throughput sequencing technologies. As key regulatory factors, circRNAs can modulate microRNAs (miRNAs) through a “sponge adsorption” mechanism, thereby influencing the post-transcriptional modification of critical genes. Given their high structural stability and widespread distribution in vivo, circRNAs have emerged as ideal candidate molecules for biomarkers and therapeutic targets in gastric cancer. This review focuses on the mechanisms by which circRNAs, through sponging miRNAs, regulate key nodes in therapeutic targets and senescence signaling pathways in gastric cancer.
-
-
-
Pediatric Diffuse High-Grade Gliomas: A Comprehensive Review Of Advanced Methods Of Diagnosis And Treatment
Available online: 30 June 2025More LessGlioblastoma multiforme (GBM) is a complex and aggressive brain tumor that presents significant diagnostic and therapeutic challenges in both adults and children. Understanding the pathogenesis, molecular biology, symptom presentation, and imaging features of GBM is vital for effective therapy. This review summarizes current knowledge on pediatric GBM, specifically Pediatric Diffuse High-Grade Gliomas (pHGG), focusing on diagnosis and treatment. GBM typically arises from the cerebral hemispheres, with gross features marked by heterogeneous morphology and aggressive cell populations. Recent advances in genomic research have shed light on distinct molecular pathways associated with primary and secondary GBMs. Clinical symptoms vary widely but commonly include neurological deficits and increased intracranial pressure. Magnetic resonance imaging (MRI), with its excellent soft tissue contrast, is crucial for diagnosing and monitoring GBM. Emerging techniques, such as diffusion-weighted imaging (DWI) and perfusion-weighted imaging (PWI), provide insights into the tumor's microstructure and vascularity, assisting in the development of therapeutic strategies and response assessment. Despite advances in imaging, challenges remain in accurately diagnosing and managing pediatric GBM due to its molecular heterogeneity and unique biological behavior. New therapeutic approaches, including targeted therapies and immunotherapy, offer hope for improving outcomes in children with GBM. Clinical trials are ongoing to evaluate these treatments alongside standard options, such as surgery, radiotherapy, and chemotherapy, to meet the unmet needs of pediatric oncology. A multidisciplinary approach, tailored to the individual characteristics of both the patient and the tumor, is essential to optimize treatment and outcomes for pediatric GBM patients. This review highlights the role of advanced MRI techniques in diagnosis, treatment, and monitoring while emphasizing the need for further research and clinical trials to develop more effective therapies for this devastating disease. Recent studies indicate a median survival rate of 12-18 months for pediatric GBM, with treatment response varying based on molecular subtypes. Clinical trials show that IDH-wild-type tumors exhibit poorer prognosis, whereas targeted therapies are improving outcomes in select patient groups.
-
-
-
An Overview of the Potential Use of Selective Serotonin Reuptake Inhibitors (SSRIs) in Cancer Treatment
Available online: 23 June 2025More LessCancer is a major health problem and the second leading cause of death worldwide. Chemotherapy remains the mainstay therapeutic option to treat cancer patients, which consists of conventional, hormonal, and/or targeted therapies. However, the significant adverse effects, negative impact on patients’ quality of life, and high costs of some medications, as well as the challenges associated with developing new drugs, are prompting the scientific community to seek innovative and alternative treatment strategies. One such strategy is drug repurposing, the use of existing drugs, already approved for other medical conditions for cancer treatment, leveraging their known safety and toxicity profiles. Among these groups are the selective serotonin reuptake inhibitors (SSRIs) that target serotonin transporter (SERT). In this review, we presented the mechanism of action of SSRIs on the systems biology level, along with their network pharmacology related to protein-protein interactions. We also showed the association of SSRIs and SERT with various diseases, including several types of cancer. Knowing the expression of SERT in cancer and being a target for SSRIs, studies have been investigating the repurposing of SSRIs for cancer treatment. This review also presents a summary of several clinical and preclinical studies that have investigated the use of SSRIs either as single agents or in combination with conventional chemotherapy for cancer treatment, showing promising results. Collectively, they have shown the antiproliferative and growth inhibition effects on cancer cells and/or tumors. We also presented the mechanism(s) of action and pathways these drugs are acting in cancer, along with molecular changes in cellular proteins and enzymes.
-