bims-meproc Biomed News
on Metabolism in Prostate Cancer
Issue of 2025–11–09
29 papers selected by
Grigor Varuzhanyan, UCLA



  1. Cell Rep. 2025 Nov 05. pii: S2211-1247(25)01301-4. [Epub ahead of print]44(11): 116530
      A high-fat diet (HFD) promotes tumor progression and therapeutic resistance, but its mechanistic role in prostate cancer (PCa) remains unclear. In this study, we show that an HFD not only accelerates PCa progression but also significantly reduces sensitivity to CDK4/6 inhibitors. Mechanistically, an HFD activates CDK4, inducing RB1 phosphorylation and facilitating E2F1 release. Meanwhile, phosphorylation of RB1 at the S249/T252 site enhances its interaction with ETS1 and suppresses ETS1's transcriptional activity. Treatment with CDK4/6 inhibitors induces dephosphorylation at this site, relieving ETS1 suppression and promoting PCYT2 expression and phosphatidylcholine metabolic reprogramming. The resulting metabolic products further disrupt RB1-E2F1 binding, leading to additional E2F1 release and increased resistance to CDK4/6 inhibitors. In conclusion, our results identify a diet-metabolism-transcriptional regulatory axis centered on RB1 phosphorylation and ETS1 reactivation, reveal a mechanism of acquired resistance to CDK4/6 inhibitors of castration-resistant PCa, and provide a theoretical basis for combinatorial strategies targeting metabolic and oncogenic signals.
    Keywords:  CDK4/6 inhibitors; CP: cancer; CP: metabolism; RB1; high-fat diet; prostate cancer
    DOI:  https://doi.org/10.1016/j.celrep.2025.116530
  2. Investig Clin Urol. 2025 Nov;66(6): 559-568
       PURPOSE: Prostate cancer that progresses despite androgen deprivation therapy, known as metastatic castration-resistant prostate cancer (mCRPC) is notoriously difficult to treat. While abiraterone acetate (AA) is a standard therapy, repurposing drugs like silodosin, a selective α1A-adrenoceptor antagonist, offers promising potential.
    MATERIALS AND METHODS: PC-3 human prostate cancer cells were treated with silodosin, AA, or their combination. Cell viability was assessed via CCK-8 assay, apoptosis by Annexin V-FITC/PI flow cytometry, and drug interaction using CompuSyn to determine combination index (CI) and dose reduction index (DRI).
    RESULTS: Both silodosin and AA reduced cell viability concentration-dependently (half-maximal inhibitory concentration [IC₅₀]: 44.16 mM and 66.90 µM, respectively). Combination therapy yielded the lowest viability (18.03%±5.73%), significantly outperforming AA (41.79%±13.11%) and silodosin (45.35%±11.51%) alone. Silodosin induced greater apoptosis (80.94%±15.88%) than AA (22.93%±7.41%), while the combination further enhanced apoptosis (87.41%±12.25%). At ¼ IC₅₀ doses, the combination retained efficacy using markedly reduced concentrations-16.72 µM AA (8-fold reduction) and 11.04 mM silodosin (5-fold reduction). CI values across fractional effects remained <1, confirming synergism, with favorable DRI values indicating enhanced efficacy at lower doses.
    CONCLUSIONS: Silodosin exhibits potent effects in reducing cell viability and promoting apoptosis in androgen-insensitive prostate cancer cells, comparable to AA. When combined, both agents demonstrate synergistic effects with reduced dosing requirements. These findings support further investigation of silodosin as a potential therapeutic in mCRPC.
    Keywords:  Abiraterone acetate; In vitro techniques; Prostatic neoplasms; Silodosin
    DOI:  https://doi.org/10.4111/icu.20250310
  3. Cancer Res. 2025 Nov 03. 85(21): 4045-4046
      Resistance to androgen deprivation therapy remains a major clinical challenge in prostate cancer, necessitating alternative therapeutic strategies. The androgen receptor (AR) plays a central role in driving resistance through mechanisms including enhanced AR signaling, DNA repair, and autophagy. In this issue of Cancer Research, Cordova and colleagues identify a metabolic vulnerability in AR protein synthesis, showing that alternate-day fasting (ADF) impairs AR translation by inducing ribosome collisions on AR mRNA. This stress response activates kinases such as p38 MAPK and selectively reduces AR protein levels, independent of transcription or protein stability. Notably, ADF enhances the efficacy of enzalutamide across multiple prostate cancer models. Unlike conventional AR-targeting approaches, ADF reveals a posttranscriptional dependency of AR expression under nutrient stress while sparing most other proteins. These findings highlight a novel translational vulnerability in prostate cancer and support combining dietary interventions with AR-targeted therapies to overcome resistance and improve patient outcomes. See related article by Cordova et al., p. 4182.
    DOI:  https://doi.org/10.1158/0008-5472.CAN-25-3234
  4. Cell Biosci. 2025 Nov 04. 15(1): 152
      Cancer-associated fibroblasts (CAFs) exhibit heterogeneity and play diverse roles in prostate cancer (PCa) progression, yet their specific impact via lactate metabolism remains unexplored. This study identifies a CAFs lactate metabolism-associated transcriptomic signature (CLS) through multi-omics analysis and establishes a Lactate Metabolism-Related Clinical Prognostic Index (LMCAFCPI) using machine learning. Lactate metabolism genes from MsigDB, were used to derive CLS from single-cell RNA sequencing (scRNA-seq). Unsupervised clustering divided samples into two risk subgroups, and GSVA and ssGSEA assessed biological functions and immune features. The LMCAFCPI demonstrated superior prognostic performance. A nomogram incorporating Gleason grade, PSA, T-stage, and LMCAFCPI showed stable biochemical recurrence (BCR) prediction. The study also revealed positive correlations between LMCAFCPI and specific immune cells, and validated signature genes in cellular models. CUT-TAG sequence confirmed PA2G4 knockdown in CAFs suppresses PCa proliferation and metastasis by inhibit H3K18la. Overall, LMCAFCPI offers a novel prognostic tool for PCa management and highlights potential targets for future research, with PA2G4 emerging as a potential therapeutic target due to its inhibition of H3K18la in PCa.
    Keywords:  CAFs; Lactate metabolism; PCa; Predictive signature
    DOI:  https://doi.org/10.1186/s13578-025-01485-9
  5. Oncol Res. 2025 ;33(11): 3493-3522
       Objectives: Despite the fact that prostate cancer is one of the most common tumors in men, this study intends to evaluate the predictive significance of immune and metabolic genes in prostate cancer using multi-omics data and experimental validation.
    Methods: The research developed and validated a prognostic model utilizing The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases, integrating immune and metabolic gene sets. Additionally, the prognostic gene Adenylate Kinase 5 (AK5) was analyzed in prostate cancer tissue microarrays from Ruijin Hospital. The functional role of the AK5 gene was validated through knockdown and overexpression experiments in four prostate cancer cell lines, employing cell proliferation assays, colony formation assays, and both xenograft models in nude mice and patient-derived xenograft models.
    Results: This research developed a prognostic model comprising ten genes, which was validated across multiple datasets for its predictive efficacy. Experimental results indicated that AK5 is significantly expressed in prostate cancer and facilitates tumor proliferation; knockdown of AK5 inhibited cell colony formation and growth of subcutaneous xenografts in nude mice, while AK5 inhibitors significantly reduced tumor volume in patient-derived xenografts.
    Conclusion: This study constructed a prognostic model with clinical potential and preliminarily confirmed the oncogenic role of AK5 in prostate cancer. The findings indicate that focusing on the immunological metabolic axis and the AK5 gene may offer novel approaches for prostate cancer treatment.
    Keywords:  immune; metabolism; prognostic model; prostate cancer
    DOI:  https://doi.org/10.32604/or.2025.066783
  6. Front Genet. 2025 ;16 1680674
      Prostate cancer poses a serious burden on men's quality of life. Identifying novel biomarkers for therapeutic development and prognostic prediction has long been a focal point in prostate cancer research. HSP family is a group of molecular chaperones that exhibit close relationship with many cancer types. In this study we screened out HSPB8 as a potential biomarker using WGCNA. Then we analyzed its expression patterns, investigated its biological functions, and assessed its prognostic values with a combination of bioinformatic analyses and experimental validation. Our data demonstrated that HSPB8 exhibited lower expression levels in prostate cancer tissues than in normal prostatic tissues. As a tumor suppressor gene, lack of HSPB8 was associated with unfavorable survival outcomes among patients with prostate cancer. In terms of biological function, HSPB8 were predominantly enriched in muscle-related biological processes, such as muscle contraction and muscle cell differentiation. On the molecular and cellular levels, HSPB8 silencing induced cellular proliferation and enhanced invasive and migratory capacities of prostate cancer cell lines. Its tumor-suppressive function was likely mediated through inactivation of PI3K-AKT signaling. Overall, this study offers a new understanding into the pathogenesis of prostate cancer, proposing that targeting HSPB8 might be a promising area in prostate cancer treatment.
    Keywords:  AKT-mTOR signaling; HSPB8; WGCNA; prostate cancer; survival-related gene
    DOI:  https://doi.org/10.3389/fgene.2025.1680674
  7. Discov Oncol. 2025 Nov 07. 16(1): 2060
       BACKGROUND: Peiminine, an active alkaloid, has been reported to exhibit antitumor properties in several malignancies. This study aims to examine the role and potential mechanism of peiminine in prostate cancer (PCa).
    METHODS: CCK-8, wound healing, colony formation, and Transwell assays were employed to evaluate PCa cell phenotypes. Cell cycle progression and apoptosis were examined by flow cytometry. Mitochondrial ROS, mitochondrial membrane potential, and ATP levels were measured to evaluate mitochondrial function in PCa cells. Western blotting was used to assess protein levels associated with apoptosis, EMT, and Wnt/β-catenin signaling. Immunofluorescence staining was performed to detect β-catenin expression. The in vivo effects of peiminine were evaluated using a xenograft mouse model.
    RESULTS: Peiminine dose-dependently impaired PCa cell viability without significantly affecting non-tumor cells. Peiminine inhibited PCa cell growth and motion and triggered apoptosis, cell cycle arrest, and mitochondrial dysfunction in vitro. Peiminine reduced PCa cell-derived tumor growth in the xenograft mouse model. Peiminine inhibited Wnt/β-catenin signal transduction.
    CONCLUSION: Peiminine exhibits an antitumor role in PCa by targeting Wnt/β-catenin signaling.
    Keywords:  Aggressiveness; Peiminine; Prostate cancer; Wnt/β-catenin pathway
    DOI:  https://doi.org/10.1007/s12672-025-03455-6
  8. Neoplasia. 2025 Nov 01. pii: S1476-5586(25)00131-9. [Epub ahead of print]71 101251
      Neuroendocrine prostate cancer (NEPC) represents a highly aggressive, treatment‑refractory phenotype that frequently emerges after androgen‑deprivation therapy (ADT). Although perturbed calcium signaling has been implicated in prostate cancer bone metastasis, the specific molecular mechanisms governing NEPC progression remain incompletely characterized. Here, we delineate the MCTP1/FYN/MEF2C signaling axis as a pivotal modulator of intracellular calcium homeostasis that drives neuroendocrine differentiation (NED) and enhances tumor aggressiveness. We demonstrate that ADT upregulates MCTP1, a transmembrane protein with calcium-sensing capabilities, which subsequently activates the Src-family kinase FYN to initiate oncogenic signaling cascades. This activation induces transcriptional upregulation of bone morphogenesis-related genes, including MEF2C and ALPL. Mechanistically, calcium-responsive transcription factors ZEB1 and ZEB2 directly transactivate MEF2C, thereby integrating calcium flux with epithelial-to-mesenchymal transition (EMT) programs in prostate cancer. Elevated ZEB1/ZEB2-dependent MEF2C expression reinforces the MCTP1/FYN kinase pathway, potentiating neuroendocrine lineage commitment and ALPL enzymatic activity. Chromatin immunoprecipitation coupled with transcriptomic analyses reveals that MEF2C directly occupies regulatory elements of MCTP1, FYN, and ALPL, enabling their calcium-dependent transcriptional activation. Structure-based virtual screening identified a potent small-molecule antagonist targeting MCTP1, which markedly attenuates tumor burden, ALPL activity, and neuroendocrine marker expression in prostate cancer in vitro and in vivo models. Collectively, these findings establish MCTP1 as a novel therapeutically exploitable vulnerability in therapy-induced NEPC, providing critical insights into the calcium-dependent oncogenic signaling networks mediated by the MCTP1/FYN/MEF2C axis in advanced prostate cancer.
    Keywords:  ALPL; Androgen deprivation therapy; FYN; MCTP1; MEF2C; Neuroendocrine differentiation; Prostate cancer
    DOI:  https://doi.org/10.1016/j.neo.2025.101251
  9. Cell Death Discov. 2025 Nov 06. 11(1): 507
      Cancer-associated fibroblasts (CAFs) are central architects of the prostate cancer (PCa) microenvironment, yet their phenotypic diversity and druggable vulnerabilities remain largely uncharted. Here, we present an integrative multi-omics characterization of primary ex vivo CAFs from seven treatment-naïve PCa patients. Using single-cell RNA sequencing (scRNA-seq), we uncover substantial transcriptional heterogeneity among CAFs, with distinct gene expression programs related to extracellular matrix remodeling, inflammation, immune modulation, and metabolic reprogramming. This phenotypic diversity was further supported by variable expression of canonical stromal markers, including FAP, SULF1, VIM, CAV1, and αSMA. Transcription factor network analysis revealed SOX, FOX, and STAT3 family members as key regulators of pro-tumorigenic CAF states. To probe therapeutic vulnerabilities, we performed high-throughput drug sensitivity and resistance testing (DSRT) across 396 oncology compounds. CAFs exhibited broad sensitivity to multikinase inhibitors, with dasatinib, midostaurin, and FGFR inhibitors (AZD4547, erdafitinib) emerging as top stromal-directed candidates. These findings underscore the plasticity of prostate CAFs and reveal actionable vulnerabilities, supporting the development of targeted stromal therapies to disrupt tumor-stroma interactions in PCa.
    DOI:  https://doi.org/10.1038/s41420-025-02792-3
  10. J Antibiot (Tokyo). 2025 Nov 07.
      Breast tumor kinase (Brk) is an intracellular kinase that initiates a downstream oncogenic signaling through phosphorylation of focal adhesion kinase (FAK) and signal transducer and activator of transcription 3 (STAT3). Demethylmycemycin A (DA) was isolated from a sponge-derived Dactylosporangium sp. OK1079. Though known as a microbial dibenzoxazepinone, its biological activity has never been investigated. Previous studies on the marine triterpene sipholenol A identified its perhydrobenzoxepine system as the key pharmacophore that mediated its Brk binding. The bioisosteric similarity of DA to the sipholenol A perhydrobenzoxepine motivated a molecular docking simulation of DA for potential Brk binding. The antiproliferative effect of DA was investigated against diverse prostate cancer (PC) cell lines including LNCaP (castration/hormone-sensitive primary adenocarcinoma), PC3, and 22Rv1 (castration-resistant), in addition to the androgen-independent DU145 cells. LNCaP cells were the most sensitive to the effects of DA, followed by PC3, showing IC50 values of 7.6 and 9.8 μM, respectively. DA treatments significantly reduced the migration and clonogenicity of the LNCaP cells. Western blot analysis indicated the ability of DA to reduce the expression levels of activated Brk, FAK and STAT3 in a dose-dependent manner in both cell lines. DA also decreased the expression levels of the total FAK but didn't affect the total level of Brk while the expression level of total STAT3 was only suppressed in LNCaP cells. These results highlight the PC proliferation and migration suppressive effects of DA through targeting Brk-FAK-STAT3 axis. DA is a potential prototype hit that can be developed particularly for Brk-expressing PC control.
    DOI:  https://doi.org/10.1038/s41429-025-00871-2
  11. Eur J Med Res. 2025 Nov 07. 30(1): 1090
       BACKGROUND: Castration-resistant prostate cancer (CRPC) represents the terminal stage of prostate cancer (PCa), yet the molecular mechanisms driving its development remain unclear. Members of the histone lysine demethylase (KDM) family regulate histone methylation and thereby modulate transcriptional programs during malignant progression, contributing to PCa pathogenesis. While the function of KDM3B in PCa has been described, its involvement in CRPC remains uncertain. This study investigated the mechanistic role of KDM3B in CRPC progression.
    METHODS: Clinical specimens and publicly available datasets were analyzed to assess KDM3B expression in PCa and CRPC tissues. Cellular proliferation was evaluated through CCK-8 and colony formation assays. In addition, RT-PCR, WB, and CCK-8 assays were employed to elucidate the relationship between KDM3B activity and CRPC development.
    RESULTS: KDM3B expression was markedly reduced in both PCa and CRPC samples. Functional assays indicated that KDM3B suppressed the proliferative capacity of CRPC cells in vivo. Moreover, KDM3B upregulated PTEN expression, and its regulatory effect on CRPC cell proliferation was mediated through PTEN modulation.
    CONCLUSION: The findings suggest that KDM3B suppresses CRPC cell proliferation by enhancing PTEN expression, highlighting its potential role as a tumor-suppressive factor in PCa.
    Keywords:  Castration-resistance PCa (CRPC); Cell proliferation; KDM3B; PTEN; Prostate cancer (PCa)
    DOI:  https://doi.org/10.1186/s40001-025-03371-z
  12. Mol Carcinog. 2025 Nov 02.
      Prostate cancer (PCa) is one of the most common genitourinary malignancies in men worldwide. As a 5-methylcytosine (m5C) methyltransferase, NSUN2 has been implicated in regulating PCa progression. This study aimed to investigate the role of NSUN2 in PCa and elucidate its underlying mechanisms. The biological behaviors of PCa cells were assessed using Cell Counting Kit-8, EdU incorporation, and Transwell assays. The expression levels of relevant RNAs were determined via quantitative real-time PCR. The interaction between NSUN2 and YES proto-oncogene 1 (YES1) was examined through methylated RNA immunoprecipitation (MeRIP), RNA immunoprecipitation (RIP), and dual-luciferase reporter assays. Results showed that NSUN2 was elevated in PCa, and its downregulation suppressed cell viability, proliferation, migration, and invasion. Mechanistically, NSUN2 interacted with YES1 and stabilized its mRNA by promoting m5C modification on YES1. The oncogenic role of NSUN2 was further confirmed in xenograft models in vivo. In conclusion, our study demonstrated that NSUN2 facilitated malignant proliferation and migration of PCa cells by enhancing YES1 mRNA stability via m5C modification. These findings suggested that both NSUN2 and YES1 may serve as potential therapeutic targets for PCa, offering new strategies for treatment.
    Keywords:  NSUN2; YES1; m5C modifications; prostate cancer
    DOI:  https://doi.org/10.1002/mc.70060
  13. Eur J Pharmacol. 2025 Nov 05. pii: S0014-2999(25)01089-1. [Epub ahead of print] 178335
      Prostate cancer (PCa) pathogenesis involves complex interactions between genetic susceptibility and exposure to endocrine-disrupting chemicals (EDCs). This study aimed to systematically identify key genes linking EDC exposure to PCa using an integrated bioinformatics and machine learning (ML) approach. We analyzed four transcriptomic datasets (GSE32571, GSE71016, GSE46602, GSE200879) and identified 437 differentially expressed genes (DEGs). By integrating these with high-confidence EDC-interacting genes from the Comparative Toxicogenomics Database (CTD), we pinpointed 169 core candidates. An ensemble ML framework, evaluating over 70 algorithm combinations, identified an optimal model (glmBoost+RF) that refined this list to eight core genes, including Glutathione S-Transferase Pi 1 (GSTP1). Mendelian randomization (MR) analysis established a causal, protective role for GSTP1 against PCa risk (OR=0.880, 95% CI=0.777-0.998, P=0.046). Molecular docking and dynamics simulations revealed stable binding between GSTP1 and high-priority EDCs, such as Benzo[a]pyrene (binding energy: -9.8 kcal/mol), indicating a direct interaction mechanism. Functional enrichment analyses implicated these genes in oxidative stress response and xenobiotic metabolism. Furthermore, single-cell RNA sequencing and immune infiltration analysis suggested a role for GSTP1 in modulating the tumor microenvironment. Our findings elucidate a critical "EDC-GSTP1-PCa" axis, highlighting GSTP1's potential as a therapeutic target and providing mechanistic insights into environmental chemical-induced prostate carcinogenesis.
    Keywords:  Endocrine-disrupting chemicals; GSTP1; Machine learning; Mendelian randomization; Molecular docking; Prostate cancer
    DOI:  https://doi.org/10.1016/j.ejphar.2025.178335
  14. Front Oncol. 2025 ;15 1673513
      Ammonium metabolism represents a critically understudied yet pivotal driver of prostate tumorigenesis and tumor microenvironment (TME) remodeling. The interplay between tumor metabolic reprogramming and the tumor microenvironment has emerged as a critical frontier in oncology research. While previous studies on prostate cancer metabolism have predominantly focused on lipid metabolism and the Warburg effect, the role of ammonium metabolism, particularly the urea cycle in tumor immune regulation remains insufficiently explored. This metabolic reprogramming constitutes a central node connecting catabolic nutrient breakdown to anabolic biosynthesis by integrating upstream amino acid deamination and transamination reactions with downstream pathways, generating key intermediates including α-ketoglutarate, coenzyme A, and citrate that concurrently fuel the tricarboxylic acid cycle and macromolecular synthesis. Crucially, oncogenic drivers such as Myc and p53 modulate this flux through epigenetic regulation of core enzymes such as glutaminase, glutamine synthetase and ornithine transcarbamylase, thereby channeling metabolism toward tumor progression. The immunomodulatory consequences manifest through dual mechanisms including TME immunosuppression driven by M2 macrophage polarization and immune evasion mediated via glutathione dependent redox homeostasis disruption. Beyond its established role in modulating redox homeostasis, ammonium metabolic reprogramming may additionally trigger novel cell death modalities such as ferroptosis by GSH/GPX4 axis. This emerging pathway offers promising therapeutic avenues for prostate cancer intervention. Synthesizing mechanistically validated insights from in vivo or in vitro models and clinical trials of ammonium-targeting inhibitors, this review proposes novel therapeutic strategies and candidate biomarkers. Moreover, the unique citrate and polyamine metabolism characteristics of prostate cancer may be impacted by these processes, offering promising avenues for future treatments.
    Keywords:  ADT; SLC; TME; ammonium metabolism; prostate cancer
    DOI:  https://doi.org/10.3389/fonc.2025.1673513
  15. J Ethnopharmacol. 2025 Nov 01. pii: S0378-8741(25)01537-5. [Epub ahead of print]356 120845
       ETHNOPHARMACOLOGICAL RELEVANCE: The Al Madina Al Munawwara variety of Phoenix dactylifera L. (Ajwa dates) is a date fruit of cultural, religious, and medicinal significance. Traditionally used in prophetic and alternative medicine, Ajwa dates are reported to possess anti-inflammatory, antioxidant, and anti-cancer properties. However, the role of Ajwa dates in targeting prostate cancer metastasis remains underexplored.
    AIM OF THE STUDY: To evaluate the anti-metastatic and anti-tumor properties of the ethyl acetate fraction of Ajwa dates (EAFAD) against prostate cancer stem-like cells (PC3) in vitro and in vivo, and to identify its bioactive compounds using Ultra-Performance Liquid Chromatography (UPLC).
    MATERIALS AND METHODS: PC3 cells were cultured in ultra-low attachment plates to enrich stem-like cells, confirmed by chemoresistance and toluidine blue staining. EAFAD was evaluated for cytotoxicity (MTT assay), anti-migratory activity (wound-healing, migration, and invasion assays), and anti-tumor efficacy using a xenograft mouse model. UPLC analysis identified bioactive compounds in EAFAD.
    RESULTS: Cancer stem-like cells were enriched, and tumorspheres were formed. EAFAD inhibited the proliferation, migration, and invasive properties of stem-like PC3 cells. The in vivo model showed suppressed tumor growth in the EAFAD-treated group, and histopathological analysis confirmed tumor cell apoptosis. The UPLC analysis of EAFAD confirmed the presence of compounds, including methyl 4-acetoxybenzoate, 2-hydroxycinnamic acid, diosmin, and isorhamnetin-3-O-glucoside, some of which are known for their anticancer and anti-inflammatory activities.
    CONCLUSION: EAFAD exhibited potent in vitro anti-proliferative effects against stem-like PC3 cells. It also demonstrated anti-migratory, anti-invasive effects and notable in vivo anti-tumor activity. These results indicate that EAFAD could serve as a potential therapeutic agent for prostate cancer.
    Keywords:  Ajwa dates; Metastasis; Phoenix dactylifera L; Prostate cancer; Stem-like cells; UPLC profiling
    DOI:  https://doi.org/10.1016/j.jep.2025.120845
  16. Cell Death Discov. 2025 Nov 06. 11(1): 502
      Neuroendocrine prostate cancer (NEPC) is an aggressive and therapy-resistant subtype of prostate cancer characterized by high levels of endoplasmic reticulum (ER) stress and metabolic dysregulation. The subsequential metabolic adaptations in the cancer cells reinforce survival mechanisms that contribute to therapy resistance and metastasis. The oncogenic driver neuroblastoma-derived MYC (MYCN) exacerbates ER stress by increasing calcium ion efflux from the ER into mitochondria, promoting glycolytic and oxidative stress. Here, we demonstrate that nitric oxide (NO) signaling is dysregulated in NEPC, thus allowing impaired S-nitrosylation of MYCN and uncontrolled ER stress. We show that exogenous NO supplementation restores MYCN S-nitrosylation at Cys4, Cys186, and Cys464. This re-establishment significantly reduces ER stress markers, inhibits the unfolded protein response (UPR), and suppresses NEPC cell proliferation and colony formation in vitro. In an orthotopic NEPC murine model, NO treatment led to a substantial reduction in tumor burden and metastasis to the liver and brain, with corresponding decreases in chromogranin and synaptophysin expression. Additionally, NO supplementation attenuated glycolytic stress by limiting calcium-mediated mitochondrial dysfunction and modulating metabolic pathways. Our findings uncover a direct mechanistic link between MYCN-driven ER stress and NEPC progression and highlight NO supplementation as a potential therapeutic strategy to counteract lineage plasticity and metabolic adaptations in NEPC. These results provide a compelling rationale for further investigation into NO-based therapies as a novel intervention for NEPC, a cancer subtype with limited treatment options and poor prognosis.
    DOI:  https://doi.org/10.1038/s41420-025-02774-5
  17. JCO Oncol Pract. 2025 Nov 07. OP2500506
       PURPOSE: Men with prostate cancer face considerable cardiovascular (CV) comorbidity. Androgen deprivation therapy (ADT) and androgen receptor pathway inhibitors (ARPIs) elevate this risk, yet no prostate-specific CV monitoring tools exist. Clinicians rely on general calculators such as the American Heart Association (AHA)/American College of Cardiology (ACC) atherosclerotic cardiovascular disease (ASCVD) score, which remain unvalidated in this setting. The purpose of this study is to evaluate changes in the estimated CV risk of men with prostate cancer after 6 months of hormone therapy using clinical tools established for the general population.
    METHODS: This was a post hoc analysis of sixty-three men with localized high-risk prostate cancer treated with 6 months of preoperative ADT plus apalutamide with or without abiraterone on a trial (ClinicalTrials.gov identifier: NCT03279250). ASCVD risk and metabolic syndrome (MetS) Z scores were calculated at baseline and the end of treatment using AHA/ACC and MetS calculators.
    RESULTS: After 6 months of ADT plus ARPI, the median ASCVD risk score increased modestly (+0.95), with 21% exhibiting a clinically significant increase (≥2.5%). This was primarily driven by increases in total cholesterol (median, 182-211) and low-density lipoprotein (median, 101-122), whereas blood pressure did not have a clinically significant change. Five patients experienced a major adverse CV event, yet only one had a clinically significant increase in ASCVD risk score. Only 28% of patients experienced an increase in MetS risk.
    CONCLUSION: After 6 months of ADT plus ARPI, men with prostate cancer showed worsening lipid profiles and 21% had worsening of their CV risk based on general population tools. These findings highlight the unmet need to develop prostate cancer-specific CV risk assessment tools.
    DOI:  https://doi.org/10.1200/OP-25-00506
  18. Appl Biochem Biotechnol. 2025 Nov 08.
      Emerging evidence underscores the pivotal yet context-dependent role of butyrate metabolism in oncogenic progression; however, the mechanistic landscape of its associated genetic determinants in prostate carcinogenesis remains poorly characterized. This gap presents a critical opportunity to delineate novel metabolic checkpoints that may offer therapeutic vulnerabilities for advanced prostate malignancies. Analyzing prostate cancer (PC) patients from TCGA and GEO databases, we identified 320 BMGs and stratified tumors into two butyrate metabolism-associated clusters. Machine learning and single-cell transcriptome analysis are used for further study. Luciferase reporter assay, qRT-PCR, FISH, and functional assays are applied to investigate the role of FOS. BMC1 correlated with aggressive phenotypes and stromal-rich tumor microenvironments, while BMC2 was linked to cell cycle regulation and DNA repair. A machine learning-derived RSF + GBM prognostic model demonstrated robust predictive accuracy for biochemical recurrence (training C-index: 0.85). BM scores are further associated with tumor mutation burden and differential drug sensitivities. FOS is overexpressed in PC, promotes proliferation and migration via transcriptional suppression of tumor-suppressive miR-27b. Clinical cohorts confirmed FOS's correlation with advanced T stages and recurrence risk. These findings establish BM-based stratification as a prognostic tool and implicate the FOS-miR-27b axis as a therapeutic target, bridging metabolic heterogeneity with molecular mechanisms in PC progression. Our research offers a critical opportunity to delineate novel metabolic checkpoints that may offer therapeutic vulnerabilities for advanced prostate malignancies.
    Keywords:  Butyrate Metabolism; FOS; Identification; MiR-27b; Prostate cancer
    DOI:  https://doi.org/10.1007/s12010-025-05476-x
  19. J Pharm Pharmacol. 2025 Nov 03. pii: rgaf100. [Epub ahead of print]
       BACKGROUND: Late-stage prostate cancer is treated with hormonal therapy. While initially effective, development of drug resistance is common. Hypoxia, a local-environmental occurrence in tumours, is known to trigger hormone-independence and concurrent drug resistance in cancer cells.
    METHODS: Here we analyse single-cell transcriptomes of LNCaP cells throughout drug treatment. These cells were exposed to chronic hypoxia and treated with Enzalutamide, a hormonal drug which inhibits the androgen receptor, both with and without Tazemetostat, an epigenetic drug that inhibits EZH2 catalytic activity, which renders Enzalutamide-resistant clones partially sensitive to hormonal therapies. We identify genes characterizing the resistant clone and assess clinical relevance.
    RESULTS: We characterize a resistant cluster present with Enzalutamide treatment but not with combination therapy. The top 10 upregulated genes in this cluster included genes previously linked to resistance: DDIT3, MDM2, and CDKN1A, and one previously proposed as a pan-cancer hallmark (HSP90B1). Analysis of clinical databases showed expression of CDKN1A, POLH, and GADD54 to be significantly upregulated in association with neuroendocrine prostate cancer.
    CONCLUSION: This work characterizes at a single-cell level the Enzalutamide resistant clone and the impact of epigenetic inhibitors on resistance development. This characterization may enable the identification of resistant and non-resistant cells by their gene expression profile.
    Keywords:  cancer; drugs; epigenetic; genes; hormonal; resistance
    DOI:  https://doi.org/10.1093/jpp/rgaf100
  20. Prostate. 2025 Nov 05.
       BACKGROUND: Intraductal carcinoma of the prostate (IDC-P) is most often considered a retrograde spread of invasive prostate cancer (PCa) into prostatic ducts, and its presence is associated with a poor prognosis. The aim of our study was to evaluate the differential expression between IDC-P and the associated invasive component and the heterogeneity of expression within IDC-P foci.
    METHODS: We studied 79 cases of PCa with an intraductal component treated by prostatectomy. TMA blocks were constructed with the intraductal and invasive components and used for immunohistochemical analysis of markers involved in the cell cycle, androgen signaling, hypoxia, DNA repair, and immune checkpoints.
    RESULTS: We found a good concordance of expression between both components for ERG, PTEN, p53, and MMR genes, which nevertheless show in some cases a loss restricted to the intraductal component. The expression of Ki67, PD-L1, and GLUT1 was increased in IDP-C compared to the invasive component. Furthermore, spatial heterogeneity was observed in the intraductal component: Ki67, ERG, androgen receptor and p53 were more expressed in the periphery of the lesion, while the expression of PD-L1 and GLUT1 was restricted to the center.
    CONCLUSIONS: Our results support a relatedness between invasive PCa and IDC-P, and show increased expression of markers related to PCa aggressiveness in the intraductal component. The spatial heterogeneity within IDC-P suggests a higher degree of hypoxia in the center of the lesion. Increased PD-L1 expression and loss of expression of some MMR genes in IDC-P could lead to increased sensitivity to immunomodulatory treatments.
    Keywords:  biomarkers; immunohistochemistry; intraductal carcinoma of the prostate; prostate cancer; tissu micro‐array
    DOI:  https://doi.org/10.1002/pros.70091
  21. Mol Biol Rep. 2025 Nov 06. 53(1): 55
       BACKGROUND: Prostate cancer (PCa) is a leading cause of cancer-related mortality, with significant racial disparities in outcomes. Long non-coding RNAs (lncRNAs) MALAT1 and TUG1 are implicated in oncogenic pathways. Aberrant RNA splicing, a hallmark of cancer, is often driven by dysregulation of serine-arginine protein kinase 1 (SRPK1), a key spliceosome regulator. The relationship between lncRNAs of splicing factors in cancer pathways remains underexplored, and thus, this study aimed to elucidate the roles of MALAT1 and TUG1 lncRNAs in relation to the SRPK1 inhibitor SPHINX31 in PCa.
    METHODS: Bioinformatics tools were used to analyze interactions between` MALAT1, TUG1, relevant miRNAs, and target genes. A resazurin assay assessed PCa cell viability (PC-3 vs. HEK293) in response to SPHINX31 at 24 and 48 h. RT-qPCR was used to quantify MALAT1 and TUG1 lncRNAs expression levels.
    RESULTS: The Alamar Blue assay indicated a time-dependent reduction in PC3 cell viability with 3 µM SPHINX31, particularly at 48 h. RT-qPCR revealed significant regulation of MALAT1 and TUG1, highlighting SRPK1's role in RNA pathways critical for tumor survival. SPHINX31 induced similar cytotoxic effects in HEK293 cells, illustrating the need for selective tumor specificity. MALAT1 expression was upregulated at 24 h, followed by a decline at 48 h, indicating cumulative cellular stress in PC-3 cells.
    DISCUSSION: This study demonstrated that SRPK1 inhibition alters lncRNA expression and splicing-related events in PCa. These findings highlight SPHINX31's potential as a therapeutic agent, especially in combination with treatments like Olaparib, necessitating further optimization for selectivity and reduced off-target effects.
    Keywords:  Alternative splicing; Angiogenesis; CeRNAs; Long non-coding RNAs; MALAT1; Prostate cancer; SRPK1; TUG1
    DOI:  https://doi.org/10.1007/s11033-025-11185-9
  22. Recent Pat Anticancer Drug Discov. 2025 Oct 29.
       INTRODUCTION: Preliminary investigations into the feasibility of Carbamoyl-phosphate synthetase 2, Aspartate transcarbamoylase, and Dihydroorotase (CAD)-targeted therapies have been conducted in a limited range of cancer types in pre-clinical studies. A comprehensive exploration of the diagnostic and prognostic capabilities of CAD, along with an understanding of its underlying biological mechanisms, is needed.
    METHODS: A range of bioinformatics tools was employed to produce an extensive pan-cancer analysis of CAD expression. Experimental validation of the role of CAD in enzalutamide resistance in prostate cells was conducted. The molecular classification and drug patents of CAD were reviewed using the Worldwide Espacenet ®.
    RESULTS: Our study revealed that CAD was upregulated in tumor tissues in most cancer types. The expression of CAD was significantly different in clinical stages, pathological grades, and clinical prognosis. The highest frequency of CAD mutation was shown, but CAD mutations did not affect the clinical outcome of cancer patients. Comprehensive data across different cancer types illustrate the relationship between the expression of CAD and tumor mutation burden (TMB), microsatellite instability (MSI), and homologous recombination deficiency (HRD). Immune infiltration algorithms showed a positive link between CAD level and the prevalence of tumor-associated fibroblasts, MDSC, mast cells, and CD4+T cells. CAD level was positively linked to the immune checkpoint, suggesting a potential synergistic effect between CAD and immunotherapy. The GSEA analysis revealed that CAD expression is significantly associated with angiogenesis and epithelial-mesenchymal transition (EMT) pathways. Finally, we demonstrated that knockdown of CAD inhibits the growth of prostate cancer (PCa) cells and resistance to enzalutamide.
    CONCLUSION: This study revealed the diagnostic and prognostic potential of CAD. Notably, CAD exhibits essential functions in PCa cell proliferation and enzalutamide resistance.
    Keywords:  CAD; enzalutamide resistance; immune infiltration; prognosis; prostate cancer.
    DOI:  https://doi.org/10.2174/0115748928412596251011092220
  23. Sci Rep. 2025 Nov 03. 15(1): 38422
      Insulin-like growth factor-1 (IGF-1) is associated with prostate cancer (PCa) development and lethality and exhibits immunosuppressive properties in other models. We investigated IGF-1's tumor-intrinsic immune effects in PCa to understand mechanisms underlying its poor immunotherapy response. Transcriptional profiling of human (DU145, 22Rv1) and murine (Myc-CaP) PCa cells revealed that IGF-1 suppresses cytokine signalling, antigen processing and presentation, and additional immune regulatory pathways. We further examined the expression of components involved in cancer cell recognition and immune evasion: the antigen processing machinery and PD-L1 checkpoint. IGF-1 downregulated key elements such as transporters associated with antigen processing (TAPs), endoplasmic reticulum aminopeptidase-1 (ERAP-1), and Class I β2-microglobulin, without significantly altering Class I allele expression. These changes were associated with reduced surface presentation of Class I complexes on Myc-CaP cells, suggesting disrupted peptide transport, processing, and/or presentation. In contrast, IGF-1 upregulated the immune checkpoint CD274 (PD-L1) via IGF receptor/AKT/ERK-dependent signalling. Analysis of TCGA Firehose Legacy PCa data showed higher CD274 expression in tumors with elevated IGF1 and IGFBP5. Multiplex immunofluorescence in primary PCa confirmed increased PD-L1 in patients with high serum IGF-1, supporting its role in immune evasion. Overall, these findings reveal a novel IGF-1-driven immunosuppressive mechanism that may underlie PCa's resistance to immunotherapy.
    DOI:  https://doi.org/10.1038/s41598-025-22288-5
  24. Adv Sci (Weinh). 2025 Nov 03. e09574
      Esophageal squamous cell carcinoma (ESCC) progression is driven by androgen receptor (AR) signaling, while small nucleolar RNAs (snoRNAs), classically involved in ribosomal RNA processing, are increasingly recognized for non-classical roles in cancer. However, their function in ESCC remains unknown. This study investigates AR-regulated snoRNAs and their mechanistic contributions to ESCC pathogenesis. SNORA80B is identified as the most AR-responsive snoRNA with oncogenic activity by transcriptomic profiling. Beyond its classical role, SNORA80B stabilizes cholesterol metabolism transcripts via N⁶-methyladenosine (m6A)-YTHDC1, driving cholesterol/DHT accumulation and lipid droplets (LDs) formation. A feedforward loop is observed wherein DHT-activated AR upregulates SNORA80B, which further enhances AR signaling through cholesterol metabolic reprogramming. Clinofibrate, identified as a SNORA80B inhibitor through high-throughput screening of FDA-approved drugs, disrupts this axis and demonstrates synergistic effects with cisplatin, overcoming resistance in ESCC. The study reveals a novel non-classical function of SNORA80B in ESCC, establishing it as a key effector of AR-driven metabolic reprogramming through m⁶A-dependent regulation. The repurposing of clinofibrate demonstrates the therapeutic potential of targeting snoRNA-mediated pathways, providing both mechanistic insights and a clinically translatable strategy for ESCC treatment. These findings redefine the functional scope of snoRNAs in cancer pathogenesis.
    Keywords:  SNORA80B; androgen receptor; cholesterol metabolism; esophageal squamous cell carcinoma; m6A modification
    DOI:  https://doi.org/10.1002/advs.202509574
  25. BMC Cancer. 2025 Nov 04. 25(1): 1697
       BACKGROUND: Increasing evidence implicates the NF-κB/p65 signaling pathway in the progression of prostate cancer (PC) and the development of resistance to androgen deprivation therapy. This study aimed to investigate the potential role of UHRF1, a key epigenetic regulator, in modulating NF-κB activity, and to evaluate its association with clinicopathological features and clinical outcomes in PC patients.
    METHODS: Differentially expressed genes (DEGs) were identified using the GSE104749 dataset. UHRF1 expression was validated across TCGA and GEO cohorts and further confirmed in clinical specimens using Western blotting and immunohistochemistry. Survival outcomes were evaluated using Kaplan-Meier and Cox regression analyses. A prognostic model was built incorporating UHRF1, Gleason score, and PSA, with validation via ROC curves and nomogram. Functional studies assessed the impact of UHRF1 silencing or overexpression on cell proliferation, apoptosis, cell cycle, glucose metabolism, and NF-κB signaling. Co-immunoprecipitation was used to assess the physical interaction between UHRF1 and p65.
    RESULTS: Higher Gleason scores, advanced clinical stage, lymph node involvement, and distant metastases were all positively connected with UHRF1 expression, which was markedly overexpressed in PC tissues. High UHRF1 expression was associated with shorter overall survival (OS) and disease-free survival (DFS), and independently predicted biochemical recurrence (BCR). A prognostic model incorporating UHRF1 achieved a high predictive accuracy (C-index = 0.752), and the corresponding nomogram demonstrated strong reliability in individualized risk assessment. In vitro, UHRF1 promoted tumor progression by enhancing proliferation, inhibiting apoptosis, driving aerobic glycolysis, and regulating the cell cycle. Mechanistically, UHRF1 bound to p65, promoted its phosphorylation, and activated NF-κB signaling.
    CONCLUSION: UHRF1 contributes to prostate cancer progression by driving tumor growth, metabolic reprogramming, and NF-κB activation. It holds promise as both a prognostic biomarker and a potential therapeutic target in PC.
    DOI:  https://doi.org/10.1186/s12885-025-15091-y
  26. Prostate. 2025 Nov 04.
       BACKGROUND: To investigate the role of HSD3B1 germline variant (1245C) in hormone therapy outcomes in Chinese prostate cancer (PCa) patients.
    METHODS: A multi-center observational study was conducted enrolling 785 PCa patients who received primary androgen deprivation therapy (ADT) in China. Genotyping of germline variant and survival data were obtained, and clinical outcomes were analysed using Cox regression models.
    RESULTS: The median follow-up time was 31 months. In the entire study cohort, the HSD3B1 variant (1245C) was significantly associated with a shorter time to castration resistance after adjusting for Gleason grade group (dominant model: hazard ratio, HR = 1.62, 95% confidence interval, 95% CI: 1.10-2.40, p = 0.015; additive model: HR = 1.55, 95% CI: 1.12-2.13, p = 0.008). Subgroup analysis (n = 438) with patients receiving only ADT for HSPC revealed a more significant association between the C allele and ADT failure (dominant model: HR = 2.37, 95% CI: 1.49-3.77, p < 0.001; additive model: HR = 1.93, 95% CI: 1.34-2.79, p < 0.001). Among patients who received next-generation hormone therapy after ADT failure, the C allele was associated with poorer abiraterone response (HR = 3.02, 95% CI: 1.07-8.50, p = 0.037); however, no significant change of response from enzalutamide was observed (HR = 0.98, 95% CI: 0.27-3.51, p = 0.972).
    CONCLUSIONS: The HSD3B1 germline variant (1245C) is linked to earlier ADT failure and diminished efficacy of abiraterone but does not affect enzalutamide in the treatment of PCa patients. These findings underscore its potential as a biomarker to guide personalized treatment in PCa.
    Keywords:  HSD3B1; androgen deprivation therapy (ADT); castration resistance; germline variant; prostate cancer (PCa)
    DOI:  https://doi.org/10.1002/pros.70089
  27. Toxicol Res (Camb). 2025 Oct;14(5): tfaf150
      Extensive clinical and epidemiological studies have shown that Aristolochic acids (AA) exhibit significant nephrotoxicity, mutagenicity, and carcinogenicity. This study aimed to systematically explore the potential molecular mechanisms by which AA induce urinary system tumors using a network toxicology approach.The carcinogenic potential of AA was predicted using ProTox, ADMETlab, and admetSAR. Potential targets of AA were identified via SEA, SwissTargetPrediction, and TargetNet, and then intersected with urinary tract tumor related genes obtained from the GeneCards database to yield common targets. A protein-protein interaction network was constructed, and GO and KEGG enrichment analyses were performed to determine their functional characteristics. LASSO regression models were built using TCGA datasets for prostate cancer, clear cell renal cell carcinoma, and bladder cancer to screen survival related hub genes. Finally, molecular docking of AA with the key targets was conducted using the CB-Dock2 platform. A total of 27 overlapping targets between AA and the three urinary tract tumors were identified. Enrichment analysis indicated that these targets are significantly involved in apoptosis, inflammatory responses, and cancer related pathways such as PI3K-Akt and MAPK. The LASSO regression models exhibited good prognostic performance across all three tumor types, with CASP3 identified as a common and significant core gene. Molecular docking analysis showed that AA can stably bind to the active pocket of CASP3.CASP3 may serve as a common key target in AA-induced urinary tract tumorigenesis. These findings provide novel theoretical insights into the molecular mechanisms by which AA promote the development of urinary system cancers.
    Keywords:  Aristolochic acids; LASSO regression; Molecular docking; Network toxicology; Urinary system tumors
    DOI:  https://doi.org/10.1093/toxres/tfaf150