bims-meproc Biomed News
on Metabolism in Prostate Cancer
Issue of 2025–08–24
ten papers selected by
Grigor Varuzhanyan, UCLA



  1. Pharmacol Res. 2025 Aug 15. pii: S1043-6618(25)00340-8. [Epub ahead of print]219 107915
      Ferroptosis, an iron-dependent form of regulated cell death characterized by lipid peroxidation, has emerged as a promising vulnerability in cancer therapy. In prostate cancer (PCa), androgen receptor (AR) signaling remains a central oncogenic driver, with androgen deprivation therapy (ADT) as the standard of care. Emerging evidence suggests a bidirectional interaction between ferroptosis and AR signaling: ADT may sensitize cancer cells to ferroptosis by disrupting iron and lipid homeostasis, while ferroptosis-induced oxidative stress may in turn attenuate AR transcriptional activity. This review synthesizes current mechanistic insights into this interplay, highlighting key AR-regulated mediators of ferroptosis resistance, including MBOAT2, SLC7A11, and PEX10. We also discuss the therapeutic potential of dual-function agents, such as darolutamide and erastin, which simultaneously inhibit AR activity and induce ferroptosis, representing a novel strategy for treating castration-resistant prostate cancer (CRPC). Finally, we propose that combining ferroptosis inducers with AR-targeted therapies, guided by metabolic and redox biomarkers, may overcome therapeutic resistance and enable precision oncology approaches for advanced PCa.
    Keywords:  Androgen deprivation therapy; Androgen receptor signaling; Dual-targeted therapy; Ferroptosis; Precision oncology; Prostate cancer
    DOI:  https://doi.org/10.1016/j.phrs.2025.107915
  2. Anticancer Agents Med Chem. 2025 Aug 12.
      Prostate cancer, timosaponin A‑III, apoptosis, autophagy, reactive oxygen species, AMPK/mTOR.
    INTRODUCTION: Timosaponin A-III (TAIII) is an effective anti-tumor ingredient extracted from the rhizomes of Anemarrhena asphodeloides. However, the effect of TAIII on prostate cancer cells (PCa) and its underlying mechanisms is rarely investigated. The current study aimed to investigate the anti-tumor effect and potential mechanisms of TAIII in PCa cells.
    METHODS: The effect of TAIII on the cell proliferation of PCa was evaluated by CCK-8 assay, colony formation assay, and EDU assay. Cell apoptosis and reactive oxygen species (ROS) production were evaluated by flow cytometry. The puncta of LC3 were detected by immunofluorescence analysis. The protein levels of apoptosis, autophagy, and AMPK/mTOR pathway were assessed by western blot. Finally, a PC3 xenograft nude mouse model was constructed to determine the effect of TAIII combined with chloroquine (CQ) in vivo.
    RESULTS: Our data showed that TAIII inhibited the proliferation of PCa cells and induced ROS-dependent apoptosis. TAIII treatment dramatically promoted the formation of LC3-positive puncta, and increased the expression of LC3B-II and P62 protein. Moreover, the combination of TAIII with CQ significantly enhanced the pro-apoptosis effect of TAIII in PCa cells and the PC3 xenograft model. In addition, the activation of the AMPK/mTOR pathway and the induction of autophagy induced by TAIII were reversed by Compound C. Suppressing AMPK with Compound C enhanced the apoptosis induced by TAIII in PCa cells.
    DISCUSSION: This study establishes TAIII as a potent anti-prostate-cancer agent that kills tumor cells via ROSdriven apoptosis while simultaneously triggering cytoprotective autophagy through the AMPK-mTOR axis. However, TAIII's clinical potential awaits pharmacokinetic, bioavailability, and toxicity evaluation.
    CONCLUSION: TAIII induced ROS-mediated cell apoptosis and promoted cytoprotective autophagy via the AMPK/mTOR pathway in PCa. These findings may provide a new strategy for combining TAIII with CQ together for PCa treatment.
    Keywords:  AMPK/mTOR.; Prostate cancer; apoptosis; autophagy; reactive oxygen species; timosaponin A‑III
    DOI:  https://doi.org/10.2174/0118715206389520250805135535
  3. bioRxiv. 2025 Aug 11. pii: 2025.08.07.669104. [Epub ahead of print]
      Phenotypic plasticity is a recognized mechanism of therapeutic resistance in prostate cancer (PCa), however current knowledge of driver mechanisms and therapeutic interventions are limited. Using genetically engineered mouse models (GEMMs) devoid of Pten and Rb1, we previously demonstrated the chromatin reprogramming factor enhancer of zeste homolog 2 (EZH2) as an important regulator of alternative transcription programs promoting phenotypic plasticity. Here, using a multi-omics approach we demonstrate that EZH2 regulates multilineage cell states dependent on the RNA binding protein Tristetraprolin (TTP) that mediates RNA stability and activation of translation. Combined chemical inhibition of EZH2 and PI3K/mTORC1 resulted in superior anti-tumor activity in murine and human phenotypic plastic models and was most significant when this combination was used with castration or enzalutamide. Together, these data indicate phenotypic plasticity dependence on coordination between EZH2, TTP and mTORC1 signaling that represent novel therapeutic dependencies for this lethal PCa phenotype.
    Graphical abstract:
    Significance Statement: EZH2 plays a key role in driving cell state transitions in neuroendocrine prostate cancer (NEPC), guiding cancer cells towards a more aggressive, therapy-resistant cell type. This transformation creates a specific vulnerability, as NEPC cells become highly reliant on both EZH2 and PI3K/mTORC1-translation signaling. Targeting this dependency, we demonstrate that combining EZH2 with PI3K/mTORC1 inhibition provides effective suppression of NEPC cell growth, offering a promising therapeutic strategy for treating this challenging and aggressive prostate cancer subtype.
    DOI:  https://doi.org/10.1101/2025.08.07.669104
  4. Discov Oncol. 2025 Aug 18. 16(1): 1579
       BACKGROUND: Mitochondrial dynamics, particularly the balance between fission and fusion, play a crucial role in cancer progression, including prostate cancer, by influencing cellular metabolism and survival. MTFP1 and MTFP2 are key regulators of mitochondrial fission, and their roles in prostate cancer warrant further investigation.
    METHODS: We conducted a comprehensive bioinformatics analysis using RNA-seq data from The Cancer Genome Atlas (TCGA) and SNP data from the UK Biobank (ukb-b-13348) GWAS dataset. Differential gene expression analysis was performed using the limma package, and pathway enrichment analysis was conducted using clusterProfiler. Hub genes were ranked using the CytoHubba algorithms. MCC was prioritized due to its robustness in identifying fully connected subgraphs. Mendelian Randomization (MR) analysis was performed using the TwoSampleMR package to assess the causal relationships between identified hub genes and prostate cancer.
    RESULTS: The analysis revealed significant differential expression of MTFP1 and MTFP2 between tumor and adjacent normal tissues, with MTFP2 showing a highly significant upregulation (p-value = 7.06e-06) and an AUC of 0.698, suggesting its potential as a biomarker. In the MR analysis, several hub genes, including ANLN, CDC45, CDCA2, and KIF15, were identified as having a significant causal relationship with prostate cancer, with effect estimates ranging from - 0.03 to 0.15 and statistically significant p-values. These findings suggest that mitochondrial dynamics and related pathways play a critical role in prostate cancer pathogenesis.
    CONCLUSION: The study highlights the potential diagnostic and prognostic value of mitochondrial fission-related genes, particularly MTFP2, in prostate cancer and underscores the importance of further investigating these pathways as therapeutic targets.
    Keywords:  Cytoscape; GWAS; Hub genes; MTFP1; MTFP2; Mendelian randomization; Mitochondrial fission; Prostate cancer
    DOI:  https://doi.org/10.1007/s12672-025-03215-6
  5. Cell Oncol (Dordr). 2025 Aug 19.
       BACKGROUND: Advanced prostate cancer (PCa) displays significant genetic heterogeneity and therapy resistance, yet the role of sensory perception pathways in its progression remains unclear.
    METHODS: We performed an integrative multi-omics analysis of sensory perception-linked mRNAs and lncRNAs from TCGA and scRNA-seq data. Unsupervised consensus clustering defined three molecular subtypes (CS1-CS3). Key biomarkers were validated in patient tissues and serum. Immune and stromal infiltration were quantified using TIDE and ESTIMATE. Single-cell trajectories characterized TSC22D3-positive T cells, and NicheNet mapped ligand-receptor interactions.
    RESULTS: Three subtypes emerged, with CS1 showing the poorest prognosis, marked chemotherapy resistance, and pronounced stromal-immune crosstalk. CS1 tumors exhibited elevated B- and T-cell infiltration and increased oxidative phosphorylation in TSC22D3-positive T cells. NicheNet analysis identified the TNF-CCL20 axis as a central mediator of immunosuppressive signaling and chemoresistance in CS1.
    CONCLUSIONS: This study establishes sensory perception-associated molecular subtypes in PCa and links CS1 chemoresistance to immune microenvironment reprogramming via TNF-CCL20 signaling. These findings offer mechanistic insights into PCa progression and suggest actionable targets to overcome therapeutic resistance.
    Keywords:  Chemoresistance; Molecular subtypes; Multi-omics; Prostate cancer; Sensory perception genes; Tumor microenvironment
    DOI:  https://doi.org/10.1007/s13402-025-01099-w
  6. bioRxiv. 2025 Aug 12. pii: 2025.08.10.669191. [Epub ahead of print]
      The mevalonate pathway produces sterols and isoprenoids that support cancer cell growth, yet its broader metabolic functions remain incompletely defined. Here, we show that this pathway sustains amino acid biosynthesis by promoting mitochondrial NAD⁺ regeneration through ubiquinone-dependent electron transport. Statin-mediated inhibition of the mevalonate pathway impairs oxidative phosphorylation, lowers the NAD⁺/NADH ratio, and suppresses de novo serine and aspartate synthesis, thereby activating the GCN2-eIF2α-ATF4 amino acid deprivation response. The resulting depletion of serine-derived glycine and one-carbon units, together with reduced aspartate availability, limits purine and pyrimidine nucleotide production. Expression of the bacterial NADH oxidase LbNOX or the alternative oxidase AOX restores NAD⁺ levels and rescues statin-induced growth inhibition. These findings suggest that impaired NAD⁺ regeneration is a key mechanism contributing to the anti-proliferative activity of statins, linking the mevalonate pathway to mitochondrial electron transport- dependent control of amino acid metabolism.
    Significance: This study identifies the mevalonate pathway as a regulator of amino acid biosynthesis through mitochondrial electron transport-dependent NAD⁺ regeneration and reveals redox disruption as a key mechanism contributing to the anti-proliferative effects of statins.
    DOI:  https://doi.org/10.1101/2025.08.10.669191
  7. Genes Dis. 2025 Nov;12(6): 101689
      RAB26 is important in the regulation of membrane trafficking and cell motility. Recently, RAB26 has received increasing attention in cancer research. However, the functional role of RAB26 in prostate cancer (PCa) remains to be elucidated. Single-cell RNA sequencing data (GSE141445) analysis indicated that RAB26 was widely expressed in PCa cells, especially in luminal cells and basal cells. High RAB26 expression in patients was found to be significantly associated with advanced pathological stage, Gleason score, and poor prognosis. Furthermore, our experimental results showed that RAB26 promoted the proliferation, migration, and invasion of PCa cells, as well as influenced the stemness of PCa cells in vitro. Besides, the transcription sequence indicated that RAB26 might promote the metastatic potential of PCa by promoting epithelial-mesenchymal transition through cascades of MAPK/ERK pathways. Finally, we found that RAB26 activated TWIST1 expression and consequently induced epithelial-mesenchymal transition, based on its interaction with the TWIST1 promoter. In conclusion, RAB26 promotes the aggressive progression of PCa and stemness of tumor cells, which is an independent biomarker for the prognosis of PCa.
    Keywords:  Cancer stem cells; EMT; Prostate cancer; RAB26; TWIST1
    DOI:  https://doi.org/10.1016/j.gendis.2025.101689
  8. medRxiv. 2025 Aug 12. pii: 2025.08.11.25333316. [Epub ahead of print]
       Translational Relevance: Despite definitive local therapy, some men with prostate cancer develop biochemical recurrence (BCR-PCa) and progress to metastatic disease. Current standard-of-care, androgen receptor pathway inhibitors (ARPIs) with or without androgen deprivation therapy (ADT) carry substantial long-term morbidity. This trial tested a novel, non-hormonal approach of 5-azacitidine (AZA) plus all-trans retinoic acid (ATRA) to induce tumor cell dormancy via epigenetic reprogramming. The regimen was well tolerated, with manageable toxicities, and showed preliminary signals of delayed PSA progression and prolonged PSA doubling time in some patients, suggesting dormancy induction. One patient achieved durable disease stabilization with ATRA maintenance. These first-in-human findings indicate that epigenetic reprogramming may modulate dormancy in BCR-PCa, offering a potential strategy to delay or minimize ADT use and its toxicities. This approach highlights the translational potential of preventing or delaying overt metastases by activating dormancy pathways in early recurrent prostate cancer.
    Purpose: Biochemical recurrence (BCR) after definitive local therapy remains a major clinical challenge in prostate cancer (PCa), with heterogeneous disease trajectories and few established strategies to delay further progression without prolonged androgen deprivation. This pilot study evaluated the combination of 5-azacitidine (AZA) and all-trans retinoic acid (ATRA) to induce tumor dormancy and delay clinical progression in patients with BCR.
    Experimental Design: In a prospective, open-label, randomized, single-institution pilot trial, patients with BCR of PCa and no recent hormonal or definitive therapy received low-dose AZA and sequential ATRA. The co-primary endpoints were changes in prostate-specific antigen doubling time (PSADT) and time to next treatment (TTNT). Safety and biomarker analyses, including bone morphogenetic protein (BMP) signaling and dormancy marker NR2F1 in circulating tumor cells (CTCs), were evaluated to investigate treatment effects on minimal residual disease dormancy.
    Results: Fourteen patients were enrolled. Treatment resulted in an increase in median PSADT from 2.45 to 4.56 months. The median TTNT was 9.6 months, with 28.6% of patients experiencing TTNT over 12 months. No new safety signals were identified; adverse events were consistent with those expected for AZA and ATRA. Analysis of circulating BMP4 and BMP7 suggested that higher BMP4 levels may correlate with treatment response. Notably, all patients achieved testosterone recovery post-treatment, likely reflecting the avoidance of ongoing androgen deprivation. Across the cohort, treatment with AZA+ATRA led to a reduction in total CTC numbers and an apparent increase in the fraction of NR2F1-positive CTCs in responders, although the small cohort size limited statistical testing.
    Conclusions: The combination of AZA and ATRA was feasible and prolonged PSA kinetics in a subset of patients with BCR of PCa, with a favorable safety profile. This epigenetic approach promoting tumor dormancy presents a potential strategy to defer progression and delay the need for continuous hormonal suppression. Larger studies are warranted to validate these findings and further explore biomarkers predictive of clinical benefit.
    DOI:  https://doi.org/10.1101/2025.08.11.25333316
  9. Crit Rev Oncol Hematol. 2025 Aug 14. pii: S1040-8428(25)00283-5. [Epub ahead of print]215 104895
       BACKGROUND: Therapeutic resistance continues to pose one of major problem in effectively managing prostate cancer (PCa) is the advancement of therapeutic resistance. A key mechanism underlying this resistance is the suppression of apoptosis, often triggered by increased expression of anti-apoptotic Bcl-2 family proteins.
    OBJECTIVE: To evaluate the outcomes of combining standard prostate cancer therapies with inhibitors targeting Bcl-2 family anti-apoptotic proteins.
    SEARCH STRATEGY: A systematic literature research was screened using the PubMed database, Scopus, and EBSCOhost (Medline Ultimate) databases up to October 30, 2024. Studies investigating the combination of standard PCa therapies with Bcl-2 family anti-apoptosis inhibitors were included. Study assessment, analysing data, and quality assessment were independently conducted by four reviewers.
    RESULTS: Twelve studies fulfilled the inclusion criteria. Six employed in vitro methods, while the remaining six used both in vitro and in vivo approaches. The standard therapies evaluated included androgen deprivation (castration), anti-androgens, and chemotherapy. Most studies utilized non-selective Bcl-2 family inhibitors, including ABT-263 and ABT-737, as well as the selective Bcl-2 inhibitor such as ABT-199. However, no studies employed selective inhibitors for Bcl-xL or Mcl-1. All selected studies indicated that anti-apoptotic inhibitors not only amplified the cytotoxic efficacy of conventional treatments but also demonstrated senolytic properties, effectively mitigating therapy-induced cellular senescence.
    CONCLUSIONS: This systematic review demonstrates that inhibition of Bcl-2 family anti-apoptotic proteins can potentiate the efficacy of standard treatments for prostate cancer. These findings provide a compelling rationale for further research into targeted combination therapies to overcome therapeutic resistance in PCa.
    Keywords:  Anti-apoptosis inhibitors; Bcl-2 family; Prostate cancer; Therapeutic resistance
    DOI:  https://doi.org/10.1016/j.critrevonc.2025.104895
  10. Front Pharmacol. 2025 ;16 1634985
       Introduction: Lactylation, a post-translational modification characterized by the attachment of lactate to protein lysine residues on proteins, plays a pivotal role in cancer progression and immune evasion. However, its implications in immunity regulation and prostate cancer prognosis remains poorly understood. This study aims to systematically examine the impact of lactylation-related genes (LRGs) on prostate cancer.
    Methods: Single-cell and bulk RNA sequencing data from patients with prostate cancer were analyzed. Data were sourced from TCGA-PRAD, GSE116918, and GSE54460, with batch effects mitigated using the ComBat method. LRGs were identified from exisiting literature, and unsupervised clustering was applied to assess their prognostic siginificance. The tumor microenvironment and functional enrichment of relevant pathways were also evaluated. A prognostic model was developed using integrative machine learning techniques, with drug sensitivy analysis included. The mRNA expression profiles of the top ten genes were validated in clinical samples.
    Results: Single-cell RNA sequencing revealed distinct lactylation signatures across various cell types. Bulk RNA-seq analysis identified 56 prognostic LRGs, classifying patients into two distinct clusters with divergent prognoses. The high-risk cluster exhibited reduced immune cell infiltration and increased resistance to specific targeted therapies. A machine learning-based prognostic signature was developed, demonstrating robust predictive accuracy for treatment responses and disease outcomes.
    Conclusion: This study offers a comprehensive analysis of lactylation in prostate cancer, identifying potential prognostic biomarkers. The proposed prognostic signature provides a novel approach to personalized treatment strategies, deepening our understanding of the molecular mechanisms driving prostate cancer and offering a tool for predicting therapeutic responses and clinical outcomes.
    Keywords:  immune microenvironment; lactylation; machine learning; personalized treatment; prognostic biomarker; prostate cancer
    DOI:  https://doi.org/10.3389/fphar.2025.1634985