bims-meproc Biomed News
on Metabolism in Prostate Cancer
Issue of 2025–08–31
thirty-one papers selected by
Grigor Varuzhanyan, UCLA



  1. Cancer Res. 2025 Aug 27.
      Malonyl-CoA, a key metabolite, is not only the building block for lipogenesis, but also a critical regulator of mitochondrial fatty acid (FA) β-oxidation. Given the altered metabolic state of many cancers, malonyl-CoA may play a role in tumor development and drug resistance, especially in malignancies characterized by abnormal lipid metabolism, such as prostate cancer (PCa). Here, we showed that the levels of malonyl-CoA were increased in PCa, especially in castration-resistant prostate cancer (CRPC). Abnormal accumulation of malonyl-CoA promoted lipogenesis and regulated metabolic processes, maintaining endoplasmic reticulum (ER) homeostasis and mitochondrial function and ultimately contributing to PCa progression. Restoration of malonyl-CoA decarboxylase (MLYCD) expression activated the unfolded protein response via the consumption of malonyl-CoA. Importantly, malonyl-CoA accumulation promoted lysine malonylation in PCa. Ran K141 malonylation increased Ran activity and enhanced androgen receptor nuclear translocation and transcriptional activity, ultimately contributing to PCa development and resistance to antiandrogens. These findings highlight the function of malonyl-CoA in PCa progression by regulating metabolic processes and malonylating Ran K141, revealing that the malonyl-CoA axis might be a reliable biomarker and a potential therapeutic target in PCa.
    DOI:  https://doi.org/10.1158/0008-5472.CAN-24-4247
  2. Biomark Res. 2025 Aug 23. 13(1): 109
      The androgen receptor signaling inhibitor enzalutamide (Enz) is one the primary therapeutic drugs for advanced prostate cancer (PCa). Nevertheless, most of patients ultimately develop resistance to Enz. Through an integrated analysis of CRISPR genome-wide and kinome-wide screens, coupled with observations of elevated expression levels in Enz-resistant cell lines and PCa tumor tissues, our study identified RPS6KC1 as a novel essential gene implicated in Enz resistance. Mechanistically, our research indicates that the Warburg effect induces H3K18 lactylation, which regulates the expression of RPS6KC1 via the transcription factor P65. Elevated expression of RPS6KC1 was found to recruit PRDX3 to the mitochondria, thereby mitigating ferroptosis. These findings suggest that the H3K18la/NF-κB/RPS6KC1/PRDX3 axis is important for the development of resistance to Enz. Our results suggest that the combination of Enz with targeted RPS6KC1 inhibition or a ferroptosis inducer may represent a promising therapeutic strategy to overcome Enz resistance.
    Keywords:  Enzalutamide; Prostate cancer; RPS6KC1; scRNA-seq
    DOI:  https://doi.org/10.1186/s40364-025-00822-x
  3. Asian Pac J Cancer Prev. 2025 Aug 01. pii: 91793. [Epub ahead of print]26(8): 2855-2867
       OBJECTIVE: Metabolic reprogramming, especially lipid reprogramming, is critical in cancer progression, including prostate cancer (PCa). Androgen deprivation therapy (ADT) is commonly used to slow down tumor spread and Lipid metabolism has been linked to its resistance in PCa. This study examines the therapeutic potential of Matairesinol (MA), a plant-derived lignan, in targeting lipid reprogramming in PCa cells.
    METHODS: PC-3 cells were treated with different doses of MA and its effect was studied on cell growth and induction of apoptosis by trypan blue dye exclusion and JC-1 dye assays, respectively. The altered expression of de novo fatty acid, cholesterol biosynthesis and other associated genes were evaluated by qPCR. Changes in intracellular lipid accumulation were assessed by Nile red staining. The bioinformatics approach was used to identify the main targets of MA using DrugBank, PubChem, and BindingDB databases, and molecular docking was performed with Autodock 4.2 to predict the binding of MA.
    RESULTS: MA significantly impaired PCa cell growth and mitochondrial membrane potential, inducing apoptosis. MA modulated mRNA expression of fatty acid and cholesterol biosynthesis, lipid transport, and lipolysis-related genes, and reduced lipid accumulation. Bioinformatics analysis with DrugBank, PubChem, and BindingDB revealed the main targets of MA; and molecular docking with AutoDock 4.2 predicted MA binding and identified SHBG and DHRS4L2 as potential targets. In vitro validation confirmed that MA significantly reduced mRNA levels of SHBG and DHRS4L2 in PC-3 cell line.
    CONCLUSION: By targeting lipid metabolism, MA holds promising potential as a therapeutic agent for PCa, especially in ADT-resistant and Metastatic Castration Resistant Prostate Cancer (mCRPC) cases.
    Keywords:  Apoptosis; Bioinformatics; Lipid regulation; PC3 cell line; SHBG
    DOI:  https://doi.org/10.31557/APJCP.2025.26.8.2855
  4. Oncogene. 2025 Aug 23.
      (Yes-associated protein 1) YAP1 is frequently activated in human prostate cancers (PCa), but the underlying regulatory mechanism remains elusive. Here, we identified a novel scaffold protein HOMER3 in PCa, that can promote YAP1 activity by disrupting LATS-YAP1 phosphorylation. Mechanistically, HOMER3 overexpression in PCa facilitates the SRC kinase to phosphorylate YAP1 accompanied by counteracting LATS1-mediated YAP1 inhibition, thereby maintaining high YAP1 nuclear localization and transcriptional activity. Accordingly, HOMER3 gain-of-function in PCa cells phenocopies the effect of YAP1 activation, including cell hyperproliferation in vitro and rapid tumor growth in vivo. Additionally, transcriptome analysis revealed that CD274 is consistently upregulated in HOMER3 overexpressing PCa cells and patients, which eventually contributed to an immunosuppressive phenotype. More importantly, blocking SRC kinase-mediated YAP1 activation improved the immunotherapy-insensitive phenotypes in PCa caused by HOMER3 overexpression. Taken together, our findings define a novel kinase-substrate interactive platform for HOMER3 to orchestrate YAP1 activity in PCa. Targeting SRC-YAP1 oncogenic axis provides new insights into the therapeutic potential for PCa patients carried HOMER3 overexpression.
    DOI:  https://doi.org/10.1038/s41388-025-03548-0
  5. J Cancer. 2025 ;16(11): 3525-3536
      Background: The expression pattern and functions of CBX4 in prostate cancers remain ambiguous. This study aims to investigate the performance of CBX4 in prostate cancer progression and preliminary inquiry potential mechanisms. Methods: The GEPIA data website was utilized to evaluate the expression patterns of CBX families and their correlations with prognosis. The "clusterprofiler" package was used for GSEA analysis. Seurat and CellChat package were used to analyze the single-cell expression profiles. The RT-qPCR, western blot and IHC staining were performed to detect the expression of CBX4 in prostate cancer tissues or cell lines. The cell functional experiments were performed, including MTT, colony formation assay, Transwell assay and scratch assay. Western blot was conducted to explore the regulation of CBX4 on EMT markers and PI3K/AKT pathway markers. Results: CBX4 was significantly up-regulated at tissue and cell levels in prostate cancer. High expression level of CBX4 was closely associated with advanced stage and poor prognosis. Of note, CBX4 was observed to promote immunosuppressive tumor environment via PDGF, VEGF, WNT signaling by cell-cell communications. In vitro experiments confirmed the expression level. Cell function and western blot proved the down-regulation of CBX4 dramatically inhibited the proliferation, invasion and migration of prostate cancer cells by targeting PI3K/AKT signaling. Conclusion: CBX4 might serve as a potential oncogene in prostate cancer progression. This study provides a new target for the treatment of prostate cancer.
    Keywords:  CBX4; PI3K/AKT signaling; prostate cancer; single-cell sequencing
    DOI:  https://doi.org/10.7150/jca.115613
  6. Cell Biol Int. 2025 Aug 27.
      Total flavonoids of Hedyotis diffusa Willd (TFHDW) is an active compound extracted from Hedyotis diffusa Willd (HDW), one of the most well-known herbs possessing antitumor effects. In this study, the potential antitumor effects of TFHDW were investigated in vitro in mouse prostate cancer cells RM1 and human prostate cancer cells LNCaP and in vivo using a xenograft tumor model involving injection of RM1 cells. Upon TFHDW treatment, RM1 and LNCaP cells exhibited augmented protein expression of the protein inhibitor of activated STAT (PIAS4) and diminished activity of signal transducer and activator of transcription 3 (STAT3), along with impaired proliferative, migratory, and invasive capacities. Ectopic STAT3 expression or PIAS4 silencing in RM1 and LNCaP cells partly annulled the inhibition effect of TFHDW treatment on cell malignant phenotypes. Mechanistic studies revealed that TFHDW elevated transcriptional activity of damage-specific DNA-binding protein 2 via PIAS4/STAT3, consequently enhancing ubiquitination and degradation of androgen receptor (AR) protein. By this, TFHDW alleviated the growth of prostate cancer in vitro and in vivo. Altogether, our work uncovers new insights into the link between TFHDW and the PIAS4/STAT3/AR axis in prostate cancer. These findings may provide a novel therapeutic option for targeting the PIAS4/STAT3/AR axis in prostate cancer.
    Keywords:  androgen receptor; prostate cancer; signal transducer and activator of transcription 3; the protein inhibitor of activated STAT; total flavonoids of Hedyotis diffusa Willd; ubiquitination and degradation
    DOI:  https://doi.org/10.1002/cbin.70070
  7. ACS Nanosci Au. 2025 Aug 20. 5(4): 284-292
      Cancer is commonly caused by a gain of function in proto-oncogenes and a simultaneous loss of function in tumor suppressor genes. Advanced prostate cancer (PCa) is often linked with changes in the activity or expression of phosphatase and tensin homologue deleted on chromosome 10 (PTEN), a well-known tumor suppressor, and androgen receptor (AR), a pro-tumorigenic transcription factor. However, no therapies exist for the simultaneous correction of tumorigenic promotion and suppressor depletion. Here, we report that concurrent PTEN restoration and AR silencing by lipid nanoparticle (LNP) delivery of PTEN messenger RNA (mPTEN) and AR small interfering RNA (siAR) elicited synergistic therapeutic effects in PCa cells. We screened various LNP formulations for the optimal delivery of both RNAs. In C4-2 and LNCaP cells, both of which are AR-positive and PTEN-null PCa cell lines, the combinatorial treatment of siAR and mPTEN LNPs resulted in much stronger cytotoxicity in vitro than the treatment of either alone. Western blot analyses revealed concurrent regulation of phosphatidylinositol 3-kinase-protein kinase B (PI3K-AKT) and extracellular signal-regulated kinase (ERK) pathways, leading to increased caspase-3 cleavage-mediated apoptosis. Our findings suggest that the strategy of RNA-mediated concurrent restoration of tumor suppressors and inhibition of tumorigenic drivers could lead to the more effective treatment of PCa and potentially other malignancies.
    Keywords:  combination therapy; lipid nanoparticle; mRNA; prostate cancer; siRNA
    DOI:  https://doi.org/10.1021/acsnanoscienceau.4c00066
  8. Cancer Res. 2025 Aug 21.
      Histologic transformation of prostate cancer from adenocarcinoma to neuroendocrine prostate cancer (NEPC) is associated with aggressive disease and poor prognosis. This lineage transition is accompanied by Polycomb complex 2 (PRC2)-mediated epigenetic de-repression of cell-fate determining transcription factors, including prospero-homeobox 1 (PROX1). Here, we sought to functionally characterize the role of PROX1 in NEPC. An unbiased CRISPR screen in two NEPC patient-derived organoid models demonstrated high cellular dependency for PROX1. Knockout of PROX1 impeded tumor growth in NEPC models and overexpression of PROX1 promoted tumor growth and spontaneous metastasis in prostate adenocarcinoma. Transcriptomic and cistromic analyses across castration resistant adenocarcinoma and neuroendocrine models pointed to PROX1-mediated regulation of neuroendocrine-lineage transcriptional programs. Immunoprecipitation followed by mass spectrometry identified three phosphorylated sites in the DNA-binding domain of PROX1 that are critical for its stability and function. CHEK1 and CDK2 were predicted to be upstream kinases that phosphorylate PROX1, and treatment with a CHEK1 or CDK2 inhibitor reduced NEPC viability. Together, these results substantiate the role of PROX1 in NEPC and identify PROX1 phosphorylation in the DNA binding domain, which might represent a therapeutic target in NEPC.
    DOI:  https://doi.org/10.1158/0008-5472.CAN-25-0636
  9. Discov Oncol. 2025 Aug 27. 16(1): 1637
       BACKGROUND: Prostate Cancer (PCa) is one of the most common malignant tumors in men. Some patients may progress to metastatic and Castration-Resistant Prostate Cancer (CRPC), leading to increased treatment difficulty and poor prognosis. With advancements in bioinformatics and machine learning technologies, the integration of multiple databases can efficiently identify core genes associated with the occurrence of PCa. Additionally, the Tumor Microenvironment (TME) and immune cell infiltration play crucial roles in the development and therapeutic response of PCa. Investigating the interactions between core genes and the immune microenvironment will enhance the understanding of the molecular mechanisms underlying PCa and provide new avenues for precision treatment.
    METHODS: This study obtained gene expression data of prostate cancer and normal tissues from the GEO (GSE69223, GSE46602) and TCGA (TCGA-PRAD) databases. Initially, each dataset was standardized, batch effects were corrected, and differentially expressed genes (DEGs) were identified, followed by GO and KEGG enrichment analyses. Subsequently, the STRING database was utilized to construct a protein-protein interaction (PPI) network, and core modules were selected using Cytoscape software. Lasso regression and Random Forest (RF) algorithms were employed to further pinpoint core genes from the key genes. In the external validation dataset (GSE46602), the diagnostic value and expression level differences of these core genes were assessed. Combining data from TCGA-PRAD, their correlations with immune cell infiltration were analyzed. The application value in molecular pathways and potential therapeutic interventions was explored through GSEA and CTRP drug sensitivity data. Finally, immunohistochemistry and Western blot experiments were conducted to confirm the expression changes of core genes in Benign Prostatic Hyperplasia (BPH) and prostate cancer tissues, alongside correlation analyses with clinical pathological parameters.
    RESULTS: Multi-omics integrative analysis identified a total of 339 common DEGs. The PPI network and machine learning algorithms further identified four potential core genes: DKK3, SNAI2, WIF1, and FOXA1. External validation and diagnostic value assessments revealed that DKK3 and WIF1 were significantly downregulated in PCa tissues and positively correlated with higher PSA levels and Gleason scores. Immune cell infiltration analysis indicated that the downregulation of these genes was closely associated with impaired adaptive immune function and matrix remodeling, and synergistically interacted with abnormalities in the Wnt/TGF-β pathways. Further analysis using the CTRP database showed that high expression of DKK3 increased cellular sensitivity to various anti-tumor drugs, whereas high expression of WIF1 might reduce the efficacy of certain small molecule inhibitors. Immunohistochemistry and protein level detection in clinical samples confirmed that the expression levels of DKK3 and WIF1 were significantly decreased in PCa tissues and were closely associated with patients' PSA levels and Gleason scores.
    CONCLUSION: Evidence from integrated databases and clinical samples indicates that WIF1 and DKK3 are significantly downregulated in PCa. Their inactivation may accelerate tumor progression and drug resistance by relieving the negative regulation of the Wnt/TGF-β pathways and affecting the immune microenvironment. Targeting the restoration or enhancement of these two tumor suppressor genes could become a new direction for the early diagnosis and precision treatment of prostate cancer. When combined with personalized medication selection, this approach is expected to significantly improve clinical outcomes for high-risk PCa patients.
    Keywords:  Bioinformatics; DKK3; Immune infiltration; Machine learning; Prostate cancer; WIF1
    DOI:  https://doi.org/10.1007/s12672-025-03488-x
  10. Int J Med Sci. 2025 ;22(13): 3454-3463
      Background: Prostate cancer (PCa) poses a significant health burden for men, with docetaxel constituting the primary therapeutic option for patients with metastatic PCa. However, the mechanisms governing docetaxel resistance remain incompletely understood. Several studies have implicated the role of the extracellular matrix (ECM) stiffness in cancer drug resistance, yet the precise role of ECM stiffness in docetaxel resistance in PCa remains elusive. The aim of this study was to explore the influence of ECM stiffness on docetaxel resistance in PCa and elucidate the underlying molecular mechanisms, thereby providing novel insights into PCa treatment. Methods: Polyacrylamide gels of varying stiffness were utilized to mimic different ECM stiffness conditions. The sensitivity of PCa cells to docetaxel was evaluated using CCK-8, TUNEL staining, flow cytometry, and western blotting. RNA-seq was employed to analyze the transcriptomic effects of different ECM stiffness on PC-3 cells. Western blotting, qPCR, and siRNA were utilized to validate the regulatory role of the key gene in the sensitivity of PCa cells to docetaxel under varying stiffness conditions. Results: Our findings indicate that high ECM stiffness enhances docetaxel resistance in PCa cells by inhibiting docetaxel-induced apoptosis. This process is mediated through the integrin-related mechanotransduction pathway. Specifically, high ECM stiffness upregulates the expression of PRRX1, thereby promoting docetaxel resistance in PCa cells. Conclusions: High ECM stiffness promotes docetaxel resistance in PCa, with PRRX1 identified as a pivotal gene in this process. These findings contribute to a deeper understanding of the mechanisms underlying docetaxel resistance in PCa and may inform the development of novel therapeutic strategies.
    Keywords:  ECM stiffness; PCa; docetaxel; drug resistance
    DOI:  https://doi.org/10.7150/ijms.111171
  11. Cancer Drug Resist. 2025 ;8 31
      Introduction: Prostate cancer (PCa) continues to be a significant cause of mortality among men, with treatment resistance often influenced by the complexity of the tumor microenvironment (TME). This study aims to develop an immune-centric prognostic model that correlates TME dynamics, genomic instability, and the heterogeneity of drug resistance in PCa. Methods: Multi-omics data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases were integrated, encompassing transcriptomic profiles of 554 TCGA-PRAD samples and 329 external validation samples. Immune cell infiltration was assessed using CIBERSORT and ESTIMATE. Weighted gene co-expression network analysis (WGCNA) was employed to identify immune-related modules. Single-cell RNA sequencing (ScRNA-seq) of 835 PCa cells uncovered subtype-specific resistance patterns. Prognostic models were constructed using least absolute shrinkage and selection operator (LASSO) regression and subsequently validated experimentally in PCa cell lines. Results: Two immune subtypes were identified: high-risk subgroups displayed TP53 mutations, increased tumor mutation burden (TMB), and enriched energy metabolism pathways. ScRNA-seq delineated three PCa cell clusters, with high-risk subtypes being sensitive to bendamustine/dacomitinib and resistant to apalutamide/neratinib. A 10-gene prognostic model (e.g., MUC5B, TREM1) categorized patients into high/low-risk groups with distinct survival outcomes (log-rank P < 0.0001). Validation in external datasets confirmed the robust predictive accuracy (AUC: 0.854-0.889). Experimental assays verified subtype-specific drug responses and dysregulation of key model genes. Discussion: This study establishes a TME-driven prognostic framework that connects immune heterogeneity, genomic instability, and therapeutic resistance in PCa. By pinpointing metabolic dependencies and subtype-specific vulnerabilities, our findings provide actionable strategies to circumvent treatment failure, such as targeting energy metabolism or tailoring therapies based on resistance signatures.
    Keywords:  Tumor microenvironment; drug resistance; immunotherapy; prognostic model; prostate cancer
    DOI:  https://doi.org/10.20517/cdr.2025.47
  12. Toxins (Basel). 2025 Aug 04. pii: 388. [Epub ahead of print]17(8):
      Ochratoxin A (OTA), a prevalent food contaminant, has been proposed as a potential contributor to the development of prostate cancer, although its precise mechanisms remain unclear. This study employed a comprehensive approach that integrated network toxicology, machine learning, and molecular docking to clarify the role of OTA in prostate cancer. The findings indicated that OTA interacts with 364 targets related to prostate cancer, and machine learning was employed to identify five key molecular targets as priorities (ESR1, TP53, TNF, INS, and EGFR). In conjunction with the results of a functional enrichment analysis, OTA was found to possibly facilitate cancer progression by disrupting endocrine function, activating oncogenic signaling pathways, reprogramming metabolism, and modulating the tumor microenvironment.
    Keywords:  environmental carcinogenesis; machine learning; molecular docking; network toxicology; ochratoxin A; prostate cancer
    DOI:  https://doi.org/10.3390/toxins17080388
  13. Nat Genet. 2025 Aug 26.
      Despite the high prevalence of ERG transcription factor translocations in prostate cancer, the mechanism of tumorigenicity remains poorly understood. Using lineage tracing, we find the tumor-initiating activity of ERG resides in a subpopulation of murine basal cells that coexpress luminal genes (BasalLum) and not in the larger population of ERG+ luminal cells. Upon ERG activation, BasalLum cells give rise to highly proliferative intermediate (IM) cells with stem-like features that coexpress basal, luminal, hillock and club marker genes, before transitioning to Krt8+ luminal cells. Transcriptomic analysis of ERG+ human prostate cancers confirms the presence of rare ERG+ BasalLum cells, as well as IM cells whose presence is associated with a worse prognosis. Single-cell analysis revealed a chromatin state in ERG+ IM cells enriched for STAT3 transcription factor binding sites and elevated expression of the KMT2A/MLL1 and DOT1L, all three of which are essential for ERG-driven tumorigenicity in vivo. In addition to providing translational opportunities, this work illustrates how single-cell approaches combined with lineage tracing can identify cancer vulnerabilities not evident from bulk analysis.
    DOI:  https://doi.org/10.1038/s41588-025-02289-w
  14. JCI Insight. 2025 Aug 22. pii: e187151. [Epub ahead of print]10(16):
      Cancer-associated fibroblasts (CAFs) are part of the tumor microenvironment (TME) that enable cancer cells to establish metastases, but the mechanisms of these interactions are not fully known. Herein, we identified a paracrine mechanism in which CAF-secreted asporin (ASPN) activated ErbB signaling and subsequent migration of adjacent prostate cancer cells. Our data support that ASPN bound directly to the ligand binding domain of human epidermal growth factor 3 (HER3) and induced HER2/HER3 heterodimerization and activation of the PI3K, MAPK, and calcium pathways. Genetic and therapeutic inhibition of HER2/HER3 ablated ASPN-induced signaling and migration. Clinically, ASPN was detected in the stroma of HER2/HER3-expressing human metastatic prostate cancer, supporting the clinical relevance of these findings and highlighting a potential therapeutic vulnerability. Antibody-drug conjugate (ADC) therapies designed to target HER2 (trastuzumab-deruxtecan) or HER3 (patritumab-deruxtecan) significantly diminished prostate cancer cell growth in vitro and tumor size in vivo, despite Aspn in the TME. Collectively, these findings indicate ASPN functions as a HER3 ligand to induce cellular migration, and inhibition with anti-HER2 or anti-HER3 ADC therapies highlights potential clinical utility for patients with metastatic castration-resistant prostate cancer that expresses HER2 or HER3.
    Keywords:  Cell biology; Oncogenes; Oncology; Prostate cancer; Signal transduction
    DOI:  https://doi.org/10.1172/jci.insight.187151
  15. Eur J Med Chem. 2025 Aug 15. pii: S0223-5234(25)00832-3. [Epub ahead of print]299 118067
      Advanced prostate cancer (PCa) frequently progresses to metastatic castration-resistant prostate cancer (mCRPC), a stage characterized by limited therapeutic options and dismal prognosis. Emerging evidence has underscored ferroptosis induction as a promising therapeutic strategy for advanced PCa. Here, we synthesized and identified a selenium-containing compound, 8m, which significantly suppressing tumor cell proliferation, migration, and stemness in both in vitro and in vivo models. Mechanistic investigations revealed that 8m inhibits the deubiquitinase USP30, leading to enhanced ubiquitination and degradation of GPX4, a critical regulator of ferroptosis. This study not only establishes the USP30/GPX4 axis as a novel molecular mechanism governing ferroptosis in PCa using 8m as a chemical probe but also provides a promising therapeutic strategy and lead compound for the treatment of advanced PCa.
    Keywords:  Ferroptosis; GPX4; Prostate cancer; Selenium-containing compounds; USP30
    DOI:  https://doi.org/10.1016/j.ejmech.2025.118067
  16. J Chem Inf Model. 2025 Aug 21.
      The androgen receptor (AR) is a nuclear receptor involved in regulating gene expression, maintaining the sexual phenotype, and contributing to the development of prostate cancer (PCa). The binding of agonists, such as dihydrotestosterone (DHT), triggers conformational changes in the AR, affecting coactivator interactions, and regulates downstream signaling pathways. Although AR activation depends on interactions between its ligand-binding domain (LBD) and coactivators, the precise impact of ligand binding on these interactions remains unclear. Antagonists such as apalutamide, bicalutamide, and enzalutamide inhibit AR activation and are used to treat PCa. However, their long-term effectiveness is often reduced due to mutations in AR-LBD, which can shift the AR from an antagonistic to an agonistic state, diminishing treatment efficacy. The mechanisms driving this conversion have not been fully elucidated. This study employed atomic-level investigations through molecular dynamics simulation with multiple replicas covering a total time frame of 10.5 μs, to investigate ligand induced perturbations in mutants AR_LBD, particularly focusing on conformational changes and the effect on AR-coactivator interaction. The results demonstrated that DHT, an agonist, stabilizes the activation function-2 region (AF-2), thereby promoting AR-coactivator interactions, while antagonists induce distinct changes in helix 12 that disrupt these interactions. In addition, F876L and T877A mutations in AR-LBD alter the ligand-to-coactivator allosteric pathway involving the coactivator, helix 3 (H3), helix 4 (H4), the loop between H3-H4, and helix 12 (H12), potentially converting the AR-apalutamide complex from an antagonistic to an agonistic state. The free energy decomposition calculations exhibited that AR mutant systems possess higher binding affinities than antagonistic ARs, with electrostatic interactions and conformational entropies associated with the determination of the binding free energies. The study suggests that point mutations in AR-LBD induce a shift from an antagonistic to an agonistic state by altering the AR and AF-2 structure, resulting in continuous coactivator recruitment and sustained AR activity. Through the application of a dynamic cross-correlation matrix, principal component analysis, free energy landscape computation, and structural community analysis, this research offers valuable insights into AR-coactivator interactions, paving the way for more effective treatments against castration-resistant prostate cancer.
    DOI:  https://doi.org/10.1021/acs.jcim.5c01406
  17. Int J Mol Sci. 2025 Aug 13. pii: 7800. [Epub ahead of print]26(16):
      Advanced prostate cancer frequently develops resistance to antiandrogen therapy and acquires an aggressive neuroendocrine phenotype. Antiandrogens stimulate peroxisome proliferator-activated receptor gamma (PPARG) signaling and cancer progression. Molecular iodine (I2) induces cytotoxic effects in prostate cancer cell lines and antineoplastic effects in neuroblastoma and breast cancer through the indirect activation of PPARG. We investigated the adjuvant effects of I2 and androgen deprivation in prostate cancer, as well as the role of PPARG in these projections. We used androgen-dependent and androgen-independent cell lines and TRAMP mice (transgenic adenocarcinoma of the mouse prostate) as biological models, as well as bicalutamide (Bic), enzalutamide (Enz), and charcoal-stripped fetal bovine serum (CS-FBS) as androgen deprivation models. I2 promoted cytotoxic effects, whereas in surviving cells, it stimulated the outgrowth of neurite-like projections, regulated lipid content, and reduced invasive capacity. Androgen deprivation plus I2 magnified these effects, while GW9662 (PPARG antagonist) did not block them. In vivo, I2 increased the degree of prostatic desmoplasia in the sham mice but did not amplify the stromal response or reduce the epithelial lesion score induced by castration in TRAMP. In conclusion, I2 showed anti-cancer (cytotoxic, anti-invasive) and pro-cancer (pro-neurite, lipid accumulation, desmoplasia) effects through a PPARG-independent mechanism.
    Keywords:  PPARG; TRAMP; androgen deprivation; iodine; lipids; prostate
    DOI:  https://doi.org/10.3390/ijms26167800
  18. Front Genet. 2025 ;16 1539745
       Background: Prostate cancer (PCa) is a prevalent malignancy and a leading cause of cancer-related death among men. Epithelial-mesenchymal transition (EMT) plays a crucial role in tumor progression, metastasis, and treatment. However, there are limited comprehensive studies on the EMT correlation with prognosis, tumor microenvironment, and therapeutic efficacy in PCa.
    Methods: We obtained mRNA expression profiles and clinical data of PCa samples, along with 1,011 protein-coding EMT-related genes from public databases. Functional annotation and consensus clustering were performed based on differentially expressed genes. An EMT prognostic signature (EPS) was constructed in the TCGA dataset after a series of bioinformatics analyses and validated in the GSE116918 dataset. The signature was used to explore clinicopathological features, genomic heterogeneity, the immune landscape, and therapy responses. Finally, we examined the expression of key genes in clinical specimens.
    Results: An EPS was established based on four key genes (MEN1, H2AFZ, UCKL1, and FUS). The patients were classified into low-risk and high-risk groups according to their median EPS risk scores. In both datasets, patients in the high-risk group exhibited significantly lower survival rates compared to those in the low-risk group. Furthermore, the EPS risk score proved to be an independent prognostic factor, and the prognostic nomogram based on the EPS risk score and T stage yielded high accuracy. Subsequent investigations found that the EPS risk score was correlated with both tumor mutation burden and genomic heterogeneity. Notably, the low-risk group displayed a higher proportion of tumor-infiltrating immune cells and exhibited better responses to chemotherapy and immunotherapy. As expected, the validation analysis confirmed substantial overexpression of MEN1, H2AFZ, UCKL1, and FUS in PCa tissues relative to adjacent normal prostate tissues.
    Conclusion: Our preliminary EPS represents a promising biomarker for predicting PCa prognosis and has great potential for clinical application.
    Keywords:  epithelial-mesenchymal transition; prognostic signature; prostate cancer; therapy response; tumor microenvironment
    DOI:  https://doi.org/10.3389/fgene.2025.1539745
  19. Int J Mol Sci. 2025 Aug 08. pii: 7700. [Epub ahead of print]26(16):
      Intracellular alkalosis and extracellular acidosis are two pathological features associated with malignant cells. They offer advantages in terms of invasiveness and proliferation. Extracellular acidification is the consequence of intracellular metabolic changes associated with a higher metabolic rate of cancer cells, potentially inducing dangerous intracellular acidification. To overcome this menace, malignant cells adapt themselves to export hydrogen ions. Therefore, it is reasonable that targeting intracellular alkalinization and extracellular acidification to prompt the reversal of such a pH gradient towards a condition comparable to normal, untransformed cells may represent a strategy helping to contrast malignant behavior. In the present study, we investigated in vitro, in prostate cancer cell models, the biological activity towards intracellular, extracellular and organelle pH of systems of molecules of vegetal origin. A few of these systems were shown to promote intracellular acidification in vitro, whereas others were shown to prevent extracellular acidification and promote lysosomal alkalinization in a cell type-dependent manner. This result clearly indicates that these systems may function as agents interfering with malignant cells inverted pH gradient. Further analysis would be necessary to unravel the cell type specificity of their effects, as well as their mechanism of action. Nevertheless, our proof-of-principle study provides evidence that such systems of molecules can be considered interesting agents in co-adjuvating anti-cancer therapies.
    Keywords:  extracellular pH; herbal extracts; intracellular pH; lysosomal pH; pH inversion; systems of molecules of vegetal origin
    DOI:  https://doi.org/10.3390/ijms26167700
  20. Crit Rev Oncol Hematol. 2025 Aug 20. pii: S1040-8428(25)00298-7. [Epub ahead of print]214 104910
      Metabolic rewiring is a hallmark of cancer, enabling tumor cells to proliferate rapidly and survive under adverse conditions. Fatty acid synthase (FASN), a key enzyme in de novo lipogenesis, is significantly upregulated in various cancers and is associated with poor prognosis and increased tumor aggressiveness. This review examines the crucial role of FASN in cancer metabolism and evaluates the therapeutic potential of FASN inhibitors. We explore the metabolic pathways critically regulated by FASN and outline its structure, function, and regulatory mechanisms. Overexpression of FASN occurs in cancers such as lung, colon, brain, breast, and prostate, where clinical trials have either been conducted or are ongoing. Pharmacologic inhibition of FASN disrupts lipid biosynthesis, leading to accumulation of metabolic intermediates, induction of metabolic stress, and cell cycle arrest/apoptosis in cancer cells. Denifanstat (TVB-2640), the first-in-class selective FASN inhibitor with favorable pharmacokinetic properties, has demonstrated robust antitumor activity in preclinical models and encouraging results in early-phase clinical studies. Clinical evidence suggests that FASN blockade not only impairs tumor growth but also potentiates the efficacy of existing treatments, including chemotherapy and targeted agents, thereby supporting its integration into combination regimens. Future clinical optimization will require the identification of predictive biomarkers to guide patient selection and treatment stratification.
    Keywords:  Cancer Metabolism; De Novo Lipogenesis (DNL); Denifanstat (TVB-2640); Fatty Acid Synthase (FASN) inhibitors; Lipid Biosynthesis; Metabolic Reprogramming
    DOI:  https://doi.org/10.1016/j.critrevonc.2025.104910
  21. Pharmaceuticals (Basel). 2025 Aug 12. pii: 1189. [Epub ahead of print]18(8):
      Background/Objectives: Copper levels are significantly elevated in both the sera and tumor tissues of various cancers, including prostate cancer. It has therefore been suggested that targeting the elevated copper levels with copper chelators could lead to selective cancer treatment. Thus, several classes of low molecular weight copper-coordinating lipophilic compounds, as well as the newly developed copper complexes of appropriately functionalized polymers, are being investigated as promising novel anticancer therapeutics. Particularly, metal-containing polymers, or metallopolymers, are systematically investigated as anticancer agents or as drug delivery systems. This study aims to utilize the strong copper-chelating properties of hyperbranched polyethyleneimine (PEI) to develop PEI:Cu metallopolymers and evaluate their selectivity and anticancer properties against several prostate cancer cell lines. Methods: A series of PEI:Cu complexes at PEI/Cu ratios that ensure that no free copper ions are present in the solution are prepared and investigated against a human non-cancerous cell line and three prostate cancer cell lines of increasing metastatic potential. Results: PEI:Cu derivatives are cytotoxic against the human prostate carcinoma metastatic PC3 and DU145 cell lines, even at the lowest tested concentrations of 5 μg/mL, while against the non-cancerous HEK293 cells, all metallopolymer derivatives exhibit insignificant cytotoxicity up concentrations of 50 μg/mL. Their cytotoxic effect is associated with mitochondria membrane potential loss and ROS production increase. Conclusions: Hyperbranched polyethyleneimine-coordinated copper(II) metallopolymers, at low concentrations, selectively induce cytotoxicity in metastatic prostate cancer cell lines without compromising the viability of non-cancerous embryonic kidney cells.
    Keywords:  anticancer properties; hyperbranched polymers; metal-chelating polymers; prostate cancer cells
    DOI:  https://doi.org/10.3390/ph18081189
  22. Biomolecules. 2025 Aug 08. pii: 1147. [Epub ahead of print]15(8):
      The tumor suppressor DAB2IP, a RasGAP and cytoplasmic adaptor protein, modulates signal transduction in response to several extracellular stimuli, negatively regulating multiple oncogenic pathways. Accordingly, the loss of DAB2IP in tumor cells fosters metastasis and enhances chemo- and radioresistance. DAB2IP is rarely mutated in cancer but is frequently downregulated or inactivated by multiple mechanisms. Solid experimental evidence shows that DAB2IP reactivation reduces cancer aggressiveness in tumors driven by multiple different oncogenic mutations, making this protein an interesting target for cancer therapy. Considering this evidence, we screened a drug library to identify molecules that increase DAB2IP protein levels. We employed CRISPR/Cas9 gene editing to generate two prostate cancer cell models in which endogenous DAB2IP is fused to HiBiT, a peptide tag that enables luminescence-based detection of protein levels in a sensitive and quantitative manner. Using this approach, we identified drugs able to increase DAB2IP levels. We focused our attention on thiostrepton, a natural cyclic oligopeptide antibiotic that has been reported to inhibit the survival of various cancer cell lines. Functional experiments revealed that the cancer-inhibitory effect of thiostrepton is reduced in the absence of DAB2IP, suggesting that upregulation of this protein contributes to its action. These findings encourage further development of thiostrepton for the treatment of solid cancers and unveil a novel molecular target underlying its anti-tumoral activity.
    Keywords:  AIP1; CRISPR-Cas9; FOXM1; HiBiT; Ras-GAP; high-throughput screening; prostate cancer; protein tagging; tumor suppressor genes
    DOI:  https://doi.org/10.3390/biom15081147
  23. J Cell Mol Med. 2025 Aug;29(16): e70806
      Prostate cancer (PCa) is an extremely heterogeneous cancer and is highly prevalent in the older male population. Since intra-tumour heterogeneity (ITH) commonly results in PCa chemotherapy resistance and recurrence, it is critical to explore its effects on tumour behaviour. Prognostic genes related to ITH were identified, and a signature was constructed using Cox regression analyses and multiple machine learning algorithms. Single-cell RNA sequencing data extracted from PCa and CRPC samples were analysed via sub-clustering, pseudotime, cell communication and drug sensitivity approaches to elucidate their function. The oncogenic potential of hub genes was confirmed by immunohistochemistry and cell proliferation assays. An 11-gene signature underlying a prostate cancer meta-program (PCMP) was generated by selecting an optimal combination of machine learning methods. Survival assays and multivariate Cox regression analyses conducted in multiple cohorts revealed the superior prognostic value of the PCMP signature. Functional enrichment analyses indicated that it dysregulates the cell cycle. Using trajectory and cell-cell communication analyses, we illustrated that PCMP genes exert oncogenic effects by enhancing the proliferation and oxidative phosphorylation of epithelial cells. Intra-cellular assays also demonstrated that CENPA and CKS1B had promising malignant potential. In summary, our research not only establishes the association between the PCMP signature and reveals its malignant characteristics, but also deepens our understanding of the mechanisms underlying PCa progression and ITH. It holds promise for the development of targeted therapeutic interventions, thereby offering clinical benefits to patients.
    Keywords:  cell cycle; immunotherapy; intra‐tumour heterogeneity; prostate cancer; single‐cell RNA sequencing
    DOI:  https://doi.org/10.1111/jcmm.70806
  24. Pharmaceuticals (Basel). 2025 Aug 21. pii: 1241. [Epub ahead of print]18(8):
      Background: Despite advances in prostate cancer treatment, castration-resistant prostate cancer (CRPC) remains clinically challenging due to inherent therapy resistance and a lack of durable alternatives. Although traditional Chinese medicine offers untapped potential, the therapeutic role of paeoniflorin (Pae), a bioactive compound derived from Paeonia lactiflora, in prostate cancer has yet to be investigated. Methods: Using an integrative approach (network pharmacology, molecular docking, and experimental validation), we identified Pae key targets, constructed protein-protein interaction networks, and performed GO/KEGG pathway analyses. A Pae-target-based prognostic model was developed and validated. In vitro and in vivo assays assessed Pae effects on proliferation, migration, invasion, apoptosis, and tumor growth. Results: Pae exhibited potent anti-CRPC activity, inhibiting cell proliferation by 60% and impairing cell migration by 65% compared to controls. Mechanistically, Pae downregulated SRC proto-oncogene, non-receptor tyrosine kinase (SRC) mRNA expression by 68%. The Pae-target-based prognostic model stratified patients into high- and low-risk groups with distinct survival outcomes. Organoid and xenograft studies confirmed Pae-mediated tumor growth inhibition and SRC downregulation. Conclusions: Pae overcomes CRPC resistance by targeting SRC-mediated pathways, presenting a promising therapeutic strategy. Our findings underscore the utility of network pharmacology-guided drug discovery and advocate for further clinical exploration of Pae in precision oncology.
    Keywords:  SRC; network pharmacology; paeoniflorin; prostate cancer; resistance
    DOI:  https://doi.org/10.3390/ph18081241
  25. J Cancer. 2025 ;16(11): 3450-3463
      Background: Fatty acid metabolism (FAM) is a crucial metabolic characteristic of tumor cells, playing a role in various pathological processes during tumor development. Till now, the prognostic role of FAM-related genes of prostate cancer (PCa) is far from fully investigation. Methods: The combinations of 10 machine learning algorithms were applied in this study. A reliable signature, FAM-related gene score (FAMRGs), was developed to predict the prognosis of patients with PCa. External data sets were used to verify the accuracy and robustness of the FAMRGs. Drug sensitivity analysis was used to predict the optimal drug for high-risk PCa patients. The underlying mechanism related to FAMRGs were investigated by functional enrichment analysis. A nomogram based on FAMRGs was developed for personalized prediction of patient prognosis. Results: A stable FAMRGs was construced and validated in 6 independent cohorts. FAMRGs accurately divided PCa patients into low and high risk group. FAMRGs showed stronger predictive ability compared with published prognostic signatures for PCa. Also, the androgen receptor signaling inhibitors (ARSI) treatment response predictive ability of FAMRGs was identified. Five drugs that were most suitable for patients in the high risk group of FAMRGs were screened. It was shown that FAMRGs involved in cell cycle-related pathways. The novel nomogram showed precisely predictive ability for the outcomes of patients with PCa. Conclusions: The FAMRGs can accurately predict the prognosis of PCa patients and is expected to direct the clinical treatment for PCa.
    Keywords:  ARSI therapy; biochemical recurrence; fatty acid metabolism; prostate cancer; risk stratification
    DOI:  https://doi.org/10.7150/jca.112597
  26. Proc Natl Acad Sci U S A. 2025 Sep 02. 122(35): e2427116122
      Inadequate response to androgen deprivation therapy (ADT) frequently arises in prostate cancer, driven by cellular mechanisms that remain poorly understood. Here, we integrated single-cell RNA sequencing, single-cell multiomics, and spatial transcriptomics to define the transcriptional, epigenetic, and spatial basis of cell identity and castration response in the mouse prostate. Leveraging these data along with a meta-analysis of human prostates and prostate cancer (PCa), we identified cellular orthologs and key determinants of ADT response and resistance. Our findings reveal that mouse prostates harbor lobe-specific luminal epithelial cell types distinguished by unique gene regulatory modules and anatomically defined androgen-responsive transcriptional programs, indicative of divergent developmental origins. Androgen-insensitive, stem-like epithelial populations-resembling human club and hillock cells-are notably enriched in the urethra and ventral prostate but are rare in other lobes. Within the ventral prostate, we also uncovered two additional androgen-responsive luminal epithelial cell types, marked by Pbsn or Spink1 expression, which align with human luminal subsets and may define the origin of distinct PCa subtypes. Castration profoundly reshaped luminal epithelial transcriptomes, with castration-resistant luminal epithelial cells activating stress-responsive and stemness programs. These transcriptional signatures are enriched in tumor cells from ADT-treated and castration-resistant PCa patients, underscoring their likely role in driving treatment resistance. Temporal tracking of cells will precisely map disease-associated cellular transitions, and our technical framework facilitates such interrogations. Collectively, our comprehensive cellular atlas of the mouse prostate illuminates the importance of lobe-specific contexts for PCa modeling and reveals potential therapeutic targets to counter castration resistance.
    Keywords:  androgen signalling; castration resistant prostate cancer; single-cell RNA sequencing; single-cell multiomics; spatial transcriptomics
    DOI:  https://doi.org/10.1073/pnas.2427116122
  27. Biomedicines. 2025 Aug 07. pii: 1929. [Epub ahead of print]13(8):
      Background: The involvement of the intratumoral microbiome in prostate cancer progression is becoming increasingly acknowledged. This study analyzed the microbiome of prostate cancer tissues from patients with localized prostate cancer (LPC, stages 1-2) and advanced prostate cancer (APC, stages 3-4) to determine its association with cancer progression. Methods: Paraffin-embedded tissue samples obtained during radical prostatectomy underwent 16S rRNA amplicon-based profiling. Results: The profile of the bacterial communities in LPC and APC differed remarkably. While species diversity remained stable, species richness (as determined by the ACE analysis) was significantly lower in APC, correlating with a decrease in Enhydrobacter (which is more abundant in LPC) and an increase in Lautropia (enriched in APC). The role of Lautropia in the progression of cancer was confirmed by in vitro studies employing cell lines from prostate cancer. Conclusions: These findings demonstrate the potential of microbiome-targeted interventions in the management of prostate cancer.
    Keywords:  16S rRNA amplicon-based profiling; cancer progression; intratumoral microbiome; microbiome-targeted therapy; prostate cancer
    DOI:  https://doi.org/10.3390/biomedicines13081929
  28. Apoptosis. 2025 Aug 21.
      Addressing prostate cancer, particularly in its aggressive forms, poses challenges that call for innovative treatment modalities. Our research focuses on developing a nanotechnological solution to enhance targeted cancer therapy. We have synthesized advanced nanocapsules embedded with indocyanine green (ICG) and conjugated with the Anti-hPSMAEC domain to improve specificity towards prostate cancer cells. These nanocapsules are engineered to perform dual-mode phototherapy through photothermal and photodynamic mechanisms. In vitro experiments demonstrated the nanocapsules effectively target and induce apoptosis in prostate cancer cells upon exposure to near-infrared light. Furthermore, in vivo assessments in murine models revealed excellent tumor localization and a substantial reduction in tumor volume with minimal impact on healthy tissues. This innovative approach underscores the potential of nanotechnology to transform the therapeutic landscape of prostate cancer by achieving precise targeting and reducing systemic side effects. Such nanocapsule systems' continued development and refinement may substantially improve clinical outcomes and provide promising therapeutic strategies for treating complex cancers.
    Keywords:  Anti-hPSMAEC domain-CS@ICG nanocapsules; Photodynamic therapy; Photothermal therapy; Prostate cancer; Specific targeting
    DOI:  https://doi.org/10.1007/s10495-025-02163-4
  29. Molecules. 2025 Aug 08. pii: 3316. [Epub ahead of print]30(16):
      First-line treatment for localized prostate cancer (PCa) includes radical prostatectomy (RP) for high-risk disease. However, in many cases, patients experience biochemical recurrence (BCR), heralded by rising prostate specific antigen (PSA) levels in the serum. Our goal was to identify metabolic pathways that are disrupted in BCR to determine potential targets of therapy. We conducted metabolomic analysis in prostate tissue from the tumors of 74 patients who underwent prostatectomy as treatment for localized PCa and correlated levels of metabolites with clinical and non-clinical factors. Cholesterol and triglycerides were upregulated in Hispanic vs. non-Hispanic and in obese vs. non-obese individuals, respectively. Both lipids and non-lipids were altered with increasing Gleason grades and clinical stages. High post-RP PSA (>0.1 ng/mL) indicated recurrence (p = 0.0094) and correlated with alterations in 141 metabolites including 114 lipids and 26 non-lipid molecules. The largest increase with high post-RP PSA was in 2-hydroxyglutaric acid (2-HG), a product of the tricarboxylic acid (TCA) cycle, that had previously been established as an oncometabolite in other cancers. 2-HG was highly selective and specific for high post-RP PSA (AUC = 0.8526; p = 0.0002) while Kaplan-Meier curves indicated that among patients who recurred, high 2-HG in the tumor reduced time-to-recurrence from 84 months (for those with low 2-HG) to 38 months (for those with high 2-HG). The addition of D2HG, an enantiomer of 2-HG, increased the growth rate of LNCaP and C4 cells, and also increased Akt and ERK phosphorylation. 2-HG is upregulated in PCa tumors from patients who experience high post-RP PSA indicative of recurrence. Future studies may target this metabolite to prevent recurrent disease.
    Keywords:  2-hydroxyglutarate; PSA; localized prostate cancer; phospholipids; tricarboxylic acid cycle; triglycerides
    DOI:  https://doi.org/10.3390/molecules30163316
  30. Biomedicines. 2025 Aug 01. pii: 1872. [Epub ahead of print]13(8):
      Background/Objectives: Androgen deprivation therapy (ADT) is widely used to manage prostate cancer (PC), but the emergence of treatment resistance remains a major clinical challenge. Although the GST family has been implicated in drug resistance, the specific role of GSTM5 remains poorly understood. This study investigates whether GSTM5, alone or in combination with clinical variables, can improve patient stratification based on the risk of early treatment resistance. Methods: In silico analyses were performed to examine GSTM5's role in protein interactions, molecular pathways, and gene expression. The rs3768490 polymorphism was genotyped in 354 patients with PC, classified by ADT response. Descriptive analysis and logistic regression models were applied to evaluate associations between genotype, clinical variables, and ADT response. GSTM5 expression related to the rs3768490 genotype and ADT response was also analyzed in 129 prostate tissue samples. Results: The T/T genotype of rs3768490 was significantly associated with a lower likelihood of early ADT resistance in both individual (p = 0.0359, Odd Ratios (OR) = 0.18) and recessive models (p = 0.0491, OR = 0.21). High-risk classification according to D'Amico was strongly associated with early progression (p < 0.0004; OR > 5.4). Combining genotype and clinical risk improved predictive performance, highlighting their complementary value in stratifying patients by treatment response. Additionally, GSTM5 expression was slightly higher in T/T carriers, suggesting a potential protective role against ADT resistance. Conclusions: The T/T genotype of rs3768490 may protect against ADT resistance by modulating GSTM5 expression in PC. These preliminary findings highlight the potential of integrating genetic biomarkers into clinical models for personalized treatment strategies, although further studies are needed to validate these observations.
    Keywords:  GSTM5; biomarker; castration-resistant prostate cancer
    DOI:  https://doi.org/10.3390/biomedicines13081872
  31. Pharmaceuticals (Basel). 2025 Aug 14. pii: 1197. [Epub ahead of print]18(8):
      Prostate cancer is the most common cancer diagnosed in men worldwide and is ranked as the fifth leading cause of cancer-related death in men globally. Background/Objectives: We aimed to identify the effectiveness of cytotoxic plant-derived xanthones against prostate cancer over the past ten years. Methods: Searches were performed in Google Scholar, Web of Science, Scopus and PubMed/Medline for ten years up to December 2024 using pre-defined inclusion and exclusion criteria. The published articles were assessed in accordance with the PRISMA 2020 procedure. Results: From a total number of n = 11,932 results, 9 were retained as included studies, which included 51 xanthones. Conclusions: Garcibractatin A and bracteaxanthone VII exhibited significant cytotoxic effects on human prostate cancer (PC-3 cells) [IC50 value of 2.93 and 4.8 μM] and the human normal prostatic stromal myofibroblast cell line (WPMY-1 cells) [IC50 value of 0.76 and 3.2 μM], which were more potent than the reference etoposide [(IC50 value of 10.07 μM) and (IC50 value of 12.98 μM)]. Parvifolixanthone A showed significant activity on PC-3 (IC50 of 4.65 μM), which was more potent than the reference 5-fluorouracil (IC50 of 30.59 μM); gaudichaudione H, cantleyanone A, isobractatin, isoforbesione, and neobractatin had strong cytotoxicity (IC50 values between 2.10 and 3.39 μM) as compared to etoposide (IC50 of 10.07 μM). Despite these positive outlooks, there are still several restrictions, most notably the absence of in vivo evidence in many studies and well-defined mechanisms of action for all the promising bioactive xanthones identified in this work as well as the absence of studies of their cytotoxicity on certain normal cells.
    Keywords:  cantleyanone A; garcibractatin A; gaudichaudione H; prostatic adenocarcinoma; xanthones
    DOI:  https://doi.org/10.3390/ph18081197