bims-meract Biomed News
on Metabolic reprogramming and anti-cancer therapy
Issue of 2025–04–27
28 papers selected by
Andrea Morandi, Università degli Studi di Firenze



  1. Med Oncol. 2025 Apr 23. 42(5): 178
      Chemotherapy resistance remains a major obstacle in breast cancer treatment. This study identifies DCAF13, a substrate recognition receptor of the CRL4 ubiquitin ligase complex, as a critical regulator of chemotherapy resistance through c-Myc-driven metabolic reprogramming. We found that DCAF13 expression was significantly upregulated in chemotherapy-resistant breast cancer cell lines compared to their parental counterparts. Inhibition of DCAF13 enhanced chemotherapy sensitivity, whereas its overexpression suppressed drug sensitivity. Mechanistically, DCAF13 upregulated c-Myc expression, driving metabolic reprogramming, characterized by increased glycolysis and oxidative phosphorylation. This metabolic shift promoted cell proliferation and resistance to chemotherapy. Clinically, DCAF13 expression correlated with poor prognosis in breast cancer patients, particularly in advanced stages and triple-negative breast cancer (TNBC). Our findings highlight the DCAF13-c-Myc axis as a critical mediator of chemotherapy resistance, suggesting that targeting this pathway could provide novel therapeutic strategies to overcome drug resistance in breast cancer. Further clinical studies are needed to explore the potential of DCAF13 as a therapeutic target.
    Keywords:  Breast cancer; Chemotherapy resistance; DCAF13; Metabolic reprogramming; c-Myc
    DOI:  https://doi.org/10.1007/s12032-025-02722-4
  2. Cell Death Dis. 2025 Apr 22. 16(1): 327
      FMS-like tyrosine kinase 3-internal tandem duplication (FLT3/ITD) is a common driver mutation that presents with a high leukemic burden and its impact on metabolic homeostasis remains to be further investigated. Here, we revealed that the oncogenic activation of FLT3/ITD induced upregulation of target genes of sterol regulatory element-binding proteins (SREBPs) in vivo and in acute myeloid leukemia patients. Quizartinib is a second-generation FLT3 inhibitor that selectively inhibits the activating FLT3 mutations. We demonstrated the critical role of SREBP1 degradation in conferring the response of FLT3/ITD cells to quizartinib. Mechanistically, quizartinib facilitated degradation of the precursor form of SREBP1 via the FLT3/AKT/GSK3 axis and reduced protein levels of its target gene fatty acid synthase (FASN). Lipidomics analysis by Liquid Chromatography Mass Spectrometry (LC-MS) demonstrated that inhibition of FLT3 altered global levels of phospholipids including reduction of cardiolipin, leading to subsequent loss of mitochondrial membrane potential. Pharmacological inhibition of SREBP1 or FASN sensitized FLT3/ITD leukemia cells to quizartinib. Quizartinib combined with SREBP inhibitor fatostatin or FASN inhibitor orlistat provided substantial therapeutic benefit over monotherapies in the murine FLT3/ITD leukemia model. Our results indicated the mechanistic link between FLT3/ITD and SREBP degradation and suggested the combination therapy via targeting FLT3/SREBP/FASN axis.
    DOI:  https://doi.org/10.1038/s41419-025-07661-6
  3. Cell Rep. 2025 Apr 19. pii: S2211-1247(25)00367-5. [Epub ahead of print]44(5): 115596
      Understanding the mechanisms by which oncogenic events alter metabolism will help identify metabolic weaknesses that can be targeted for therapy. Telomerase reverse transcriptase (TERT) is essential for telomere maintenance in most cancers. Here, we show that TERT acts via the transcription factor forkhead box O1 (FOXO1) to upregulate glutamate-cysteine ligase (GCLC), the rate-limiting enzyme for de novo biosynthesis of glutathione (GSH, reduced) in multiple cancer models, including glioblastoma (GBM). Genetic ablation of GCLC or pharmacological inhibition using buthionine sulfoximine (BSO) reduces GSH synthesis from [U-13C]-glutamine in GBMs. However, GCLC inhibition drives de novo pyrimidine nucleotide biosynthesis by upregulating the glutamine-utilizing enzymes glutaminase (GLS) and carbamoyl-phosphate synthetase 2, aspartate transcarbamoylase, and dihydroorotatase (CAD) in an MYC-driven manner. Combining BSO with the glutamine antagonist JHU-083 is synthetically lethal in vitro and in vivo and significantly extends the survival of mice bearing intracranial GBM xenografts. Collectively, our studies advance our understanding of oncogene-induced metabolic vulnerabilities in GBMs.
    Keywords:  CP: Cancer; CP: Metabolism; TERT; brain tumors; cancer; glioblastoma; glutamine metabolism; glutathione; in vivo stable isotope tracing; metabolic synthetic lethality; metabolomics; nucleotide biosynthesis; telomerase reverse transcriptase
    DOI:  https://doi.org/10.1016/j.celrep.2025.115596
  4. Genes Dis. 2025 Jul;12(4): 101376
      Breast cancer is the most common malignant tumor threatening women's health. Alteration in lipid metabolism plays an important role in the occurrence and development of many diseases, including breast cancer. The uptake, synthesis, and catabolism of lipids in breast cancer cells are significantly altered, among which the metabolism of fatty acids, cholesterols, sphingolipids, and glycolipids are most significantly changed. The growth, progression, metastasis, and drug resistance of breast cancer cells are tightly correlated with the increased uptake and biosynthesis of fatty acids and cholesterols and the up-regulation of fatty acid oxidation. Cholesterol and its metabolite 27-hydroxycholesterol promote the progression of breast cancer in a variety of ways. The alteration of lipid metabolism could promote the epithelial-mesenchymal transition of breast cancer cells and lead to changes in the tumor immune microenvironment that are conducive to the survival of cancer cells. While the accumulation of ceramide in cancer cells shows an inhibitory effect on breast cancer. This review focuses on lipid metabolism and elaborates on the research progress of the correlation between different lipid metabolism and the growth, progression, and drug resistance of breast cancer.
    Keywords:  Breast cancer; Ceramide; Cholesterol; Drug resistance; Fatty acids; Lipid metabolism; Tumor progression
    DOI:  https://doi.org/10.1016/j.gendis.2024.101376
  5. Anal Cell Pathol (Amst). 2025 ;2025 5714423
      Cisplatin-based chemotherapy resistance is a common issue for cancer clinical efficacy. Metformin is being studied for its possible anticancer effect. The present study aimed to investigate whether metformin affects the chemosensitivity of gastric cancer to cisplatin and reveal the molecular mechanism. In this study, the effects of combination therapy with metformin and cisplatin on cell viability, cell apoptosis, malondialdehyde, superoxide dismutase, reactive oxygen species level, glucose uptake, lactate production, protein level, and xenograft tumor formation were analyzed in gastric cancer cells. Immunohistochemical staining was performed to detect Ki67 expression in matched tumor samples. The results showed that NCI-N87 and SNU-16 cells were most resistant and sensitive to cisplatin, respectively. Metformin treatment increased the cisplatin sensitivity of gastric cancer by inhibiting cell viability and metabolic reprogramming and promoting cell apoptosis and oxidative stress. Furthermore, overexpression of nuclear factor erythroid 2-related factor 2 (Nrf2) reversed the effects of metformin in the cisplatin sensitivity of gastric cancer by inhibiting cell viability and metabolic reprogramming and promoting cell apoptosis and oxidative stress. Metformin activated p53 and AMPK pathways in cisplatin-induced NCI-N87 cells, which were reversed by upregulating Nrf2. BAY-3827 (AMPK inhibitor) or p-nitro-Pifithrin-α (p53 inhibitor) treatments also reversed the effects of metformin increased the cisplatin sensitivity of gastric cancer by inhibiting cell viability and metabolic reprogramming and promoting cell apoptosis and oxidative stress. These results suggest that metformin significantly increases chemosensitivity of gastric cancer to cisplatin by inhibiting Nrf2 expression and metabolic reprogramming and activating oxidative stress and the pathway of p53 and AMPK.
    Keywords:  AMPK; Nrf2; cisplatin resistance; gastric cancer; metformin; p53
    DOI:  https://doi.org/10.1155/ancp/5714423
  6. Nat Commun. 2025 Apr 24. 16(1): 3874
      Identification of isocitrate dehydrogenase (IDH) mutations has uncovered the crucial role of metabolism in gliomagenesis. Oncolytic herpes virus (oHSV) initiates direct tumor debulking by tumor lysis and activates anti-tumor immunity, however, little is known about the role of glioma metabolism in determining oHSV efficacy. Here we identify that oHSV rewires central carbon metabolism increasing glucose utilization towards oxidative phosphorylation and shuttling glutamine towards reductive carboxylation in IDH wildtype glioma. The switch in metabolism results in increased lipid synthesis and cellular ROS. PKC induces ACSL4 in oHSV treated cells leading to lipid peroxidation and ferroptosis. Ferroptosis is critical to launch an anti-tumor immune response which is important for viral efficacy. Mutant IDH (IDHR132H) gliomas are incapable of reductive carboxylation and hence ferroptosis. Pharmacological blockade of IDHR132H induces ferroptosis and anti-tumor immunity. This study provides a rationale to use an IDHR132H inhibitor to treat high grade IDH-mutant glioma patients undergoing oHSV treatment.
    DOI:  https://doi.org/10.1038/s41467-025-58911-2
  7. Exp Hematol Oncol. 2025 Apr 24. 14(1): 60
       BACKGROUND: Ferroptosis, a regulated cell death driven by iron-dependent lipid peroxidation, is associated with chemoresistance in lung adenocarcinoma (LUAD). This study aims to investigate the role of sarcosine in ferroptosis and its underlying mechanisms.
    METHODS: An RSL3-induced ferroptosis model was used to screen a library of 889 human endogenous metabolites and metabolomic profiling was harnessed to identify metabolites associated with ferroptosis. Cell viability, lipid-reactive oxygen species (ROS), ferrous iron, malondialdehyde (MDA), and mitochondrial integrity were assessed to evaluate sarcosine's effects on ferroptosis. Metabolic fate was studied using 15N-labeled sarcosine. Next, we used untargeted metabolomic profiling and next-generation sequencing to dissect metabolic and transcriptomic changes upon sarcosine supplementation. The effects of sarcosine on ferroptosis and chemotherapy were further validated in patient-derived organoids (PDOs), xenograft models, and LUAD tissues.
    RESULTS: Sarcosine emerged as a potent ferroptosis inducer in the metabolic library screening, which was further confirmed via cell viability, lipid-ROS, ferrous iron, and MDA measurements. Metabolic flux analysis showed limited conversion of sarcosine to other metabolites in LUAD cells, while untargeted metabolomic profiling and seahorse assays indicated a metabolic shift from glycolysis to oxidative phosphorylation. Sarcosine enhanced pyruvate dehydrogenase activity to generate more ROS by interacting with PDK4, reducing PDHA1 phosphorylation. As a co-activator of N-methyl-D-aspartate receptor (NMDAR), sarcosine also exerted its pro-ferroptosis effect via regulating ferrous export through the NMDAR/MXD3/SLC40A1 axis. Given the significance of ferroptosis in chemotherapy, we validated that sarcosine enhanced the sensitization of cisplatin by promoting ferroptosis in LUAD cells, PDOs, and xenograft models.
    CONCLUSION: Sarcosine promotes ferroptosis and enhances chemosensitivity, suggesting its potential as a therapeutic agent in treating LUAD.
    Keywords:  Chemotherapy; Ferroptosis; Lung adenocarcinoma; Organoids; Sarcosine
    DOI:  https://doi.org/10.1186/s40164-025-00657-0
  8. Cell Death Dis. 2025 Apr 23. 16(1): 331
      Acute myeloid leukemia (AML) is an aggressive hematological malignancy. Patients with wild-type FLT3 relapsed or refractory (R/R) AML face significant therapeutic challenges due to the persistent lack of effective treatments. A comprehensive understanding of the mechanisms underlying chemotherapy resistance is needed to the development of effective treatment strategies. Therefore, we investigated the molecular mechanisms underlying cytarabine (Ara-C) resistance and daunorubicin (DNR) tolerance in Ara-C-resistant RHI-1 cells derived from the wild-type FLT3 AML cell line SHI-1. Quantitative analysis of intracellular drug concentrations, proteomics, and phosphoproteomics showed that DNR resistance in Ara-C-resistant RHI-1 cells is driven by metabolic remodeling toward mitochondrial metabolism, upregulation of DNA repair pathways, and enhanced reactive oxygen species (ROS) detoxification rather than reduced drug uptake. Moreover, targeting these compensatory mechanisms, particularly the OXPHOS complex I proteins, significantly improved the efficacy of both Ara-C and DNR. Conclusively, these findings highlight mitochondrial metabolism and DNA repair as critical factors in chemotherapy resistance and offer valuable insights into potential therapeutic targets for enhancing treatment outcomes in patients with wild-type FLT3 R/R AML.
    DOI:  https://doi.org/10.1038/s41419-025-07653-6
  9. Cell Rep. 2025 Apr 19. pii: S2211-1247(25)00366-3. [Epub ahead of print]44(5): 115595
      In this study, we report that KIF26B is upregulated in bladder cancer and acts as an independent prognostic factor. Knockdown of kif26b blocks the proliferation, metastasis, and cisplatin resistance of bladder cancer cells. Mechanistically, TCF4 potently stimulates kif26b transcription by directly binding to its promoter. KIF26B activates the Wnt/β-catenin signaling pathway through association with TRAF2 and thus promotes the formation of the TCF4/β-catenin complex. KIF26B promotes the protein stability of TRAF2 by facilitating the OTUB2-mediated de-ubiquitination of TRAF2. Importantly, KIF26B promotes the nuclear translocation of TRAF2 through enhancing its association with IPO11, a process that is dependent on the C-terminal domain of β-catenin. Additionally, phosphorylation of tyrosine 78 in TRAF2 is essential for its binding to KIF26B in response to Wnt3a signaling. Furthermore, a KIF26B/TRAF2/PD-L1 axis is identified in bladder cancer, and combined therapy of anti-B7-H3 antibody with kif26b knockdown yields superior anti-tumor effects.
    Keywords:  CP: Cancer; KIF26B; TRAF2; Wnt/β-catenin signaling; bladder cancer; pathogenesis
    DOI:  https://doi.org/10.1016/j.celrep.2025.115595
  10. Cell Prolif. 2025 Apr 21. e70036
      Glutaminase-1 (GLS1) has garnered considerable interest as a metabolic target in cancer due to its heightened involvement and activity. However, the precise fate of glutaminolysis catalysed by GLS1 in cancer cells remains elusive. We found that GLS1 knockout led to significant suppression of cancer cell proliferation, which can be reversed or partially restored by supplementation of glutamate or non-essential amino acids that can be converted into glutamate. The addition of spliceosomal KGA or GAC ameliorates cancer cell growth in vitro and in vivo, providing both simultaneously completely reverse the effect. The primary metabolic fate of glutamate produced through glutaminolysis in cancer cells is mainly used to produce glutathione (GSH) for redox homeostasis, not entering the tricarboxylic acid cycle or synthesising nucleotides. GSH monoethyl ester (GSH-MEE) effectively rescues the inhibition of cancer cell proliferation caused by GLS1 knockout. Deletion of GLS1 results in an elevation of reactive oxygen species (ROS) and malondialdehyde (MDA), a reduction of NADPH/NADP+ ratio, and an augmented susceptibility of cells to ferroptosis. Glutathione Peroxidase 4 (GPX4) and GPX1 exhibit complementary roles in redox regulation, with GLS1 knockout promoting GPX4 degradation. Pharmacological inhibition of GLS1 synergises with GPX4 inhibitor to suppress tumour growth. Dual targeting of GPX4 and GPX1 presents a potent anti-cancer strategy. This metabolic mechanism facilitates a deeper comprehension of the abnormal glutamine metabolism in cancer cells, establishing a theoretical basis for the potential clinical utilisation of GLS1 inhibitors and presenting novel perspectives for advancing combinatorial therapeutic approaches.
    Keywords:  GLS1; GPX4; GSH; cancer cell; ferroptosis; glutamate
    DOI:  https://doi.org/10.1111/cpr.70036
  11. Redox Biol. 2025 Apr 15. pii: S2213-2317(25)00150-8. [Epub ahead of print]83 103637
      The hypothesis of a significant shift from oxidative phosphorylation (OXPHOS) to glycolysis in a number of solid tumors has been dominant for many years. Recently, however, evidence has begun to accumulate that OXPHOS is the major mode of energy production in many neoplasias, especially those that have undergone chemo- or radiotherapy, and especially in chemoresistant malignancies. In the present work, we demonstrated that chemoresistant triple-negative breast cancer cells prefer to obtain energy via OXPHOS to a greater extent than cells sensitive to chemotherapeutic agents, and therefore the former can be affected by some OXPHOS inhibitors. From a drug library containing several dozen antimicrobials, we selected those that inhibit OXPHOS in resistant TNBC cells and lead to mitochondrial dysfunction. We have also identified several pathways by which inhibition of growth suppression of chemoresistant cells occurs, including increased oxidative stress and mitophagy. Experiments in mice showed that selected OXPHOS inhibitors preferentially suppress tumor growth from chemoresistant but not from chemosensitive cells. The results of the present study suggest combinatorial therapy of such inhibitors and conventional anticancer drugs on resistant forms of tumors, if the latter show enhanced OXPHOS.
    Keywords:  Antimicrobials; Autophagy; Cancer resistance; Mitochondria; Oxidative phosphorylation; Triple negative breast tumors
    DOI:  https://doi.org/10.1016/j.redox.2025.103637
  12. Oncogenesis. 2025 Apr 22. 14(1): 11
      Fructose-1,6-bisphosphatase 1 (FBP1), a rate-limiting enzyme in gluconeogenesis, is important for cancer progression. The post-translational regulation of FBP1 in hypoxic environments is still unclear. Here, we report that FBP1 is down-regulated, and a low expression level of FBP1 predicts a poor prognosis in pancreatic cancer. A hypoxic environment makes FBP1 more prone to degradation, and this effect can be reversed by inhibiting global O-GlcNAcylation signalling. O-linked N-acetylglucosamine transferase (OGT) interacts with FBP1 and induces its O-GlcNAcylation at serine 47 residue (FBP1-S47) to modulate its protein function in pancreatic cancer cells. O-GlcNAcylation of FBP1-S47 promotes FBP1 degradation and also influences the expression of canonical HIF-1α target genes involved in glucose metabolism, resulting in an increase in glucose uptake and lactate secretion in pancreatic cancer cells. In addition, O-GlcNAcylation of FBP1-S47 facilitates FBP1 K48-linked polyubiquitination at lysine 51 residue (FBP1-K51), in which GlcNAc moiety can serve as a prerequisite for an FBP1 ubiquitin ligase. FBP1 (K51) K48-linked polyubiquitination mediated protein degradation can also promote cancer progression, similarly to the O-GlcNAcylation of FBP1-S47. Our data uncover a mechanism whereby FBP1 can be regulated by a protein O-GlcNAcylation-polyubiquitination axis, paving the way to cancer cell metabolic reprogramming.
    DOI:  https://doi.org/10.1038/s41389-025-00555-4
  13. Cell Commun Signal. 2025 Apr 21. 23(1): 189
      Radiotherapy is critical component of multidisciplinary cancer care, used as a primary and adjuvant treatment for patients with head and neck squamous cell carcinoma. This study investigates how mannose, a naturally occurring monosaccharide, combined with phosphomannose isomerase (PMI) depletion, enhances the sensitivity of HPV-negative head and neck tumour models to radiation. Isogenic PMI knockout models were generated by CRISPR/Cas9 gene editing, yielding a 20-fold increase in sensitivity to mannose in vitro, and causing significant tumour growth delay in vivo. This effect is driven by metabolic reprogramming, resulting in potent glycolytic suppression coupled with consistent depletion of ATP and glycolytic intermediates in PMI-depleted models. Functionally, these changes impede DNA damage repair following radiation, resulting in a significant increase in radiation sensitivity. Mannose and PMI ablation supressed both oxygen consumption rate and extracellular acidification, pushing cells towards a state of metabolic quiescence, effects contributing to increased radiation sensitivity under both normoxic and hypoxic conditions. In 3D-tumoursphere models, metabolic suppression by mannose and PMI depletion was shown to elevate intra-tumoursphere oxygen levels, contributing to significant in vitro oxygen-mediated radiosensitisation. These findings position PMI as a promising anti-tumour target, highlighting the potential of mannose as a metabolic radiosensitiser enhancing cancer treatment efficacy.
    Keywords:  Head and neck cancer; Mannose; Phosphomannose isomerase; Radiotherapy; Tumour metabolism
    DOI:  https://doi.org/10.1186/s12964-025-02204-0
  14. Cell Death Differ. 2025 Apr 23.
      Bladder cancer (BC) remains a major global health challenge, with poor prognosis and limited therapeutic options in advanced stages. TTK protein kinase (TTK), a serine/threonine kinase, has been implicated in the progression of various cancers, but its role in BC has not been fully elucidated. In this study, we show that TTK is significantly upregulated in BC tissues and cell lines, correlating with poor patient prognosis. Functional assays revealed that TTK promotes proliferation and inhibits apoptosis of BC cells. Mechanistically, TTK enhances mitophagy by directly phosphorylating ULK1 at Ser477, thereby activating the ULK1/FUNDC1-mediated mitophagy pathway. TTK knockdown disrupts mitophagy, leading to impaired clearance of damaged mitochondria, excessive accumulation of mitochondrial reactive oxygen species (mtROS), and activation of mitochondrial apoptosis. Furthermore, TTK phosphorylates SRSF3 at Ser108, preventing ULK1 exon 5 skipping and maintaining ULK1 mRNA stability. These findings show that TTK plays a key role in maintaining mitophagy in BC cells. Targeting TTK could offer a promising new approach for BC treatment by disrupting mitophagy and inducing mitochondrial apoptosis.
    DOI:  https://doi.org/10.1038/s41418-025-01492-w
  15. Sci Adv. 2025 Apr 25. 11(17): eadu0847
      Current treatments for advanced prostate cancer (PCa) primarily target the androgen receptor (AR) pathway. However, the emergence of castration-resistant prostate cancer (CRPC) and resistance to AR pathway inhibitors (APPIs) remains ongoing challenges. Here, we present BSJ-5-63, a proteolysis-targeting chimera (PROTAC) targeting cyclin-dependent kinases (CDKs) CDK12, CDK7, and CDK9, offering a multipronged approach to CRPC therapy. BSJ-5-63 degrades CDK12, diminishing BRCA1 and BRCA2 expression and inducing a sustained "BRCAness" state. This sensitizes cancer cells to PARP inhibitors (PARPis) regardless of their homologous recombination repair (HRR) status. Furthermore, CDK7 and CDK9 degradation attenuates AR signaling, enhancing its therapeutic efficacy. Preclinical studies, including both in vitro and in vivo CRPC models, demonstrate that BSJ-5-63 exerts potent antitumor activity in both AR-positive and AR-negative setting. This study introduces BSJ-5-63 as a promising therapeutic agent that addresses both DNA repair and AR signaling mechanisms, with potential benefits for a board patient population.
    DOI:  https://doi.org/10.1126/sciadv.adu0847
  16. Gut. 2025 Apr 23. pii: gutjnl-2025-335220. [Epub ahead of print]
       BACKGROUND: We conducted an integrated cross-species spatial assessment of transcriptomic and metabolomic alterations associated with progression of intraductal papillary mucinous neoplasms (IPMNs), which are bona fide cystic precursors of pancreatic ductal adenocarcinoma (PDAC).
    OBJECTIVE: We aimed to uncover biochemical and molecular drivers that underlie malignant progression of IPMNs to PDAC.
    DESIGN: Matrix-assisted laser desorption/ionisation (MALDI) mass spectrometry (MS)-based spatial imaging and Visium spatial transcriptomics (ST) was performed on human resected IPMN/PDAC tissues (n=23) as well as pancreata from a mutant Kras;Gnas mouse model of IPMN/PDAC. Functional studies in murine IPMN/PDAC-derived Kras;Gnas cells were performed using CRISPR/cas9 technology, small interfering RNAs, and pharmacological inhibition.
    RESULTS: MALDI-MS analyses of patient tissues revealed long-chain hydroxylated sulfatides to be selectively enriched in the neoplastic epithelium of IPMN/PDAC. Integrated ST analyses showed cognate transcripts involved in sulfatide biosynthesis, including UGT8, Gal3St1, and FA2H, to co-localise with areas of sulfatide enrichment. Genetic knockout or pharmacological inhibition of UGT8 in Kras;Gnas IPMN/PDAC cells decreased protein expression of FA2H and Gal3ST1 with consequent alterations in mitochondrial morphology and reduced mitochondrial respiration. Small molecule inhibition of UGT8 elicited anticancer effects via ceramide-mediated compensatory mitophagy and activation of intrinsic apoptosis pathways. In vivo, UGT8 inhibition suppressed tumour growth in allograft models of murine IPMN/PDAC cells derived from Kras;Gnas and Kras;Tp53;Gnas mice.
    CONCLUSION: Our work identifies enhanced sulfatide metabolism as an early metabolic alteration in cystic precancerous lesions of the pancreas that persists through invasive neoplasia and a potential actionable vulnerability in IPMN-derived PDAC.
    Keywords:  METABOLOMICS; PANCREATIC CANCER
    DOI:  https://doi.org/10.1136/gutjnl-2025-335220
  17. Nat Commun. 2025 Apr 24. 16(1): 3867
      The ability of cancer cells to evade immune destruction is governed by various intrinsic factors including their metabolic state. Here we demonstrate that inactivation of dihydroorotate dehydrogenase (DHODH), a pyrimidine synthesis enzyme, increases cancer cell sensitivity to T cell cytotoxicity through induction of ferroptosis. Lipidomic and metabolomic analyses reveal that DHODH inhibition reduces CDP-choline level and attenuates the synthesis of phosphatidylcholine (PC) via the CDP-choline-dependent Kennedy pathway. To compensate this loss, there is increased synthesis from phosphatidylethanolamine via the phospholipid methylation pathway resulting in increased generation of very long chain polyunsaturated fatty acid-containing PCs. Importantly, inactivation of Dhodh in cancer cells promotes the infiltration of interferon γ-secreting CD8+ T cells and enhances the anti-tumor activity of PD-1 blockade in female mouse models. Our findings reveal the importance of DHODH in regulating immune evasion through a CDP-choline dependent mechanism and implicate DHODH as a promising target to improve the efficacy of cancer immunotherapies.
    DOI:  https://doi.org/10.1038/s41467-025-59307-y
  18. Cell Death Dis. 2025 Apr 24. 16(1): 336
      Glioblastoma (GBM) is among the deadliest cancers, characterized by poor prognosis and median survival of 12-15 months post-diagnosis. Despite aggressive therapeutic regimens, GBM treatment is still an unmet clinical need due to heterogeneity, recurrencies, and resistance. Metabolic reshaping is emerging as a critical mechanism supporting cell proliferation and sustaining chemoresistance. In this study, we explored metabolic changes induced by chemotherapy in temozolomide (TMZ)-sensitive and TMZ-resistant GBM cell lines. We found that purine levels were altered in sensitive versus resistant GBM cells, highlighting a critical role of guanosine and inosine metabolism. By using a mesenchymal-like GBM zebrafish model, we uncovered dysregulated pathways involved in purine metabolism, with a downregulation of catabolic processes. Our data indicate that combined treatment with TMZ plus guanosine and inosine increased cytotoxicity, enhancing chemotherapy effectiveness in TMZ-resistant cells. These effects correlated with alterations in mitochondrial dynamics and activity. Specifically, the combinatorial effectiveness of TMZ with guanosine and inosine was linked to Mitofusin-2 overexpression, enhancing mitochondrial fusion, typically associated with a better prognosis. Therefore, our findings suggest that purine metabolism is involved in the metabolic rewiring of TMZ-resistant cells, suggesting guanosine and inosine as potential adjuvant treatments to improve the cytotoxicity effects of chemotherapy in resistant GBM.
    DOI:  https://doi.org/10.1038/s41419-025-07667-0
  19. J Exp Clin Cancer Res. 2025 Apr 24. 44(1): 129
       BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is among the cancer types with poorest prognosis and survival rates primarily due to resistance to standard-of-care therapies, including gemcitabine (GEM) and olaparib. Particularly, wild-type (wt)BRCA tumours, the most prevalent in PDAC, are more resistant to DNA-targeting agents like olaparib, restraining their clinical application. Recently, we disclosed a monoterpene indole alkaloid derivative (BBIT20) as a new inhibitor of homologous recombination (HR) DNA repair with anticancer activity in breast and ovarian cancer. Since inhibition of DNA repair enhances the sensitivity of cancer cells to chemotherapy, we aimed to investigate the anticancer potential of BBIT20 against PDAC, particularly carrying wtBRCA.
    METHODS: In vitro and in vivo PDAC models, particularly human cell lines (including GEM-resistant PDAC cells), patient-derived organoids and xenograft mice of PDAC were used to evaluate the anticancer potential of BBIT20, alone and in combination with GEM or olaparib. Disruption of the BRCA1-BARD1 interaction by BBIT20 was assessed by co-immunoprecipitation, immunofluorescence and yeast two-hybrid assay.
    RESULTS: The potent antiproliferative activity of BBIT20, superior to olaparib, was demonstrated in PDAC cells regardless of BRCA status, by inducing cell cycle arrest, apoptosis, and DNA damage, while downregulating HR. The disruption of DNA double-strand breaks repair by BBIT20 was further reinforced by non-homologous end joining (NHEJ) suppression. The inhibition of BRCA1-BARD1 heterodimer by BBIT20 was demonstrated in PDAC cells and confirmed in a yeast two-hybrid assay. In GEM-resistant PDAC cells, BBIT20 showed potent antiproliferative, anti-migratory and anti-invasive activity, overcoming GEM resistance by inhibiting the multidrug resistance P-glycoprotein, upregulating the intracellular GEM-transporter ENT1, and downregulating GEM resistance-related microRNA-20a and GEM metabolism enzymes as RRM1/2. Furthermore, BBIT20 did not induce resistance in PDAC cells. It inhibited the growth of patient-derived PDAC organoids, by inducing apoptosis, repressing HR, and potentiating olaparib and GEM cytotoxicity. The enhancement of olaparib antitumor activity by BBIT20 was confirmed in xenograft mice of PDAC. Notably, it hindered tumour growth and liver metastasis formation, improving survival of orthotopic xenograft mice of PDAC. Furthermore, its potential as a stroma-targeting agent, reducing fibrotic extracellular matrix and overcoming desmoplasia, associated with an enhancement of immune cell response by depleting PD-L1 expression in tumour tissues, renders BBIT20 even more appealing for combination therapy, particularly with immunotherapy.
    CONCLUSION: These findings underscore the great potential of BBIT20 as a novel multifaceted anticancer drug candidate for PDAC treatment.
    Keywords:  Anticancer agent; BRCA1-BARD1 interaction inhibitor; DNA damage repair; Indole alkaloid BBIT20; PDAC
    DOI:  https://doi.org/10.1186/s13046-025-03389-5
  20. Nature. 2025 Apr 23.
      Pancreatic ductal adenocarcinoma (PDAC) subsists in a nutrient-deregulated microenvironment, making it particularly susceptible to treatments that interfere with cancer metabolism1,2. For example, PDAC uses, and is dependent on, high levels of autophagy and other lysosomal processes3-5. Although targeting these pathways has shown potential in preclinical studies, progress has been hampered by the difficulty in identifying and characterizing favourable targets for drug development6. Here, we characterize PIKfyve, a lipid kinase that is integral to lysosomal functioning7, as a targetable vulnerability in PDAC. Using a genetically engineered mouse model, we established that PIKfyve is essential to PDAC progression. Furthermore, through comprehensive metabolic analyses, we found that PIKfyve inhibition forces PDAC to upregulate a distinct transcriptional and metabolic program favouring de novo lipid synthesis. In PDAC, the KRAS-MAPK signalling pathway is a primary driver of de novo lipid synthesis. Accordingly, simultaneously targeting PIKfyve and KRAS-MAPK resulted in the elimination of the tumour burden in numerous preclinical human and mouse models. Taken together, these studies indicate that disrupting lipid metabolism through PIKfyve inhibition induces synthetic lethality in conjunction with KRAS-MAPK-directed therapies for PDAC.
    DOI:  https://doi.org/10.1038/s41586-025-08917-z
  21. Adv Sci (Weinh). 2025 Apr 24. e2415983
      Numerous aberrant splicing events are implicated in tumor progression, yet comprehensive reports on splicing factors and events associated with peritoneal metastasis in gastric cancer (GCPM) are lacking. In this study, PTBP3 is found to be significantly overexpressed in peritoneal metastatic tissues of gastric cancer compared to primary tumor tissues, and higher PTBP3 expression correlates with poorer prognosis. Using gastric cancer cells and patient-derived organoids (PDO), the role of PTBP3 in promoting tumor invasion and proliferation is investigated. Mechanistically, through full-length transcriptome sequencing, PTBP3 mediates exon 4 skipping in its target gene COX11, leading to shorter transcripts that impair COX11 protein function, reducing mitochondrial copper content and enabling tumor cells to evade cuproptosis. Antisense oligonucleotide (ASO) drugs targeting the short COX11 transcripts effectively degrade mRNA, disrupting copper homeostasis. In PDO-based xenograft models, exogenous copper ionophores combined with ASO drugs induce excessive copper accumulation in mitochondria, triggering proteotoxic stress and cuproptosis. Overall, PTBP3-mediated exon 4 skipping in COX11 pre-mRNA is critical for tumor cell survival and progression in GCPM, offering potential therapeutic strategies targeting copper metabolism.
    Keywords:  alternative splicing; antisense oligonucleotide drugs; cuproptosis; gastric cancer peritoneal metastasis; patient‐derived organoids‐based xenograft
    DOI:  https://doi.org/10.1002/advs.202415983
  22. Cancer Metab. 2025 Apr 22. 13(1): 19
       BACKGROUND: Intrahepatic cholangiocarcinoma (ICC) is the second most common primary hepatocellular cancer. This study investigated whether ETV4, ALYREF, and PKM2 affect glycolytic metabolism and ferroptosis, thereby potentially influencing ICC.
    METHODS: Bioinformatic analysis was used to explore the expression levels and prognosis of ETV4, ALYREF, and PKM2 in ICC and their regulatory relationships were confirmed using in vitro experiments. Glycolytic metabolism and ferroptosis were examined, and chromatin immunoprecipitation and RNA immunoprecipitation experiments were performed to verify whether the ETV4, PKM2, and ALYREF could bind. The effect of ETV4/ALYREF on ICC was further confirmed by in vivo experiments.
    RESULTS: ETV4, ALYREF, and PKM2 were highly expressed in ICC. Overexpressed (oe)-ETV4 and oe-PKM2 promoted cell migration and increased glucose (GLU) utilization and lactate and intracellular adenosine triphosphate (ATP) production. Addition of the ferroptosis inducer Erastin to the above groups revealed that sh-ETV4 and sh-ALYREF increased lipid reactive oxygen species (ROS), malondialdehyde (MDA), and Fe2+ levels, and oe-PKM2 reversed these effects in the sh-ETV4 and sh-ALYREF groups. Oe-ETV4 promoted the expression of PKM2, whereas sh-ALYREF inhibited the same. ETV4 could bind to ALYREF and PKM2 promoter, and ALYREF could promote the stability of PKM2 in an m5C-dependent manner. In vivo, ETV4 promotes tumor growth and the expression of proteins related to glycolytic metabolism by regulating ALYREF.
    CONCLUSION: ETV4 promotes ICC development and ferroptosis resistance by facilitating glycolytic metabolism, and regulating PKM2 transcription by directly binding to the PKM2 promoter. Additionally, it mediates m5C-dependent PKM2 stabilization by directly binding to ALYREF. This study identified a new potential therapeutic target for ICC.
    Keywords:  ALYREF; ETV4; Ferroptosis; Glycolytic metabolism; ICC; PKM2; m5C
    DOI:  https://doi.org/10.1186/s40170-025-00387-1
  23. NAR Cancer. 2025 Jun;7(2): zcaf015
      Prostate cancer (PCa) is the second most common cancer worldwide and the fifth leading cause of cancer-related deaths among men. The emergence of metastatic castration-resistant prostate cancer (mCRPC) after androgen deprivation therapy (ADT) exemplifies the complex disease management for PCa. PARP inhibitors (PARPis) are being tested to treat mCRPC in tumors with defective homologous recombination repair (HRR) to address this complexity. However, increasing resistance towards PARPi in HRR-deficient patients and the low percentage of HRR-defective mCRPC patients requires the identification of new genes whose deficiency can be exploited for PARPi treatment. XRCC1 is a DNA repair protein critical in the base excision repair (BER) and single strand break repair (SSBR) pathways. We analyzed PCa patients' cohorts and found that XRCC1 expression varies widely, with many patients showing low XRCC1 expression. We created XRCC1 deficiency in PCa models to examine PARPi sensitivity. XRCC1 loss conferred hypersensitivity to PARPi by promoting the accumulation of DNA double-strand breaks, increasing cell-cycle arrest, and inducing apoptosis. We confirmed that XRCC1 expression correlated with PARPi sensitivity using a doxycycline-inducible system. Therefore, we conclude that XRCC1 expression level predicts response to PARPi, and the clinical utility of PARPi in PCa can extend to low XRCC1 expressing tumors.
    DOI:  https://doi.org/10.1093/narcan/zcaf015
  24. Int Immunopharmacol. 2025 Apr 18. pii: S1567-5769(25)00655-1. [Epub ahead of print]156 114665
      Despite the identification of several pathogenic drivers, the molecular mechanisms underlying the development of acute myeloid leukemia (AML) remain largely unknown. Therefore, we sought to explore the key genes associated with leukemia and identified cluster of differentiation 40 (CD40) as a key mediator linked to the incidence and progression of AML. Higher levels of CD40 were detected in patients with AML compared to healthy donors. Moreover, elevated CD40 expression was associated with lower overall survival rates. Furthermore, anti-CD40 antibody significantly induced apoptosis and enhanced drug sensitivity in human AML cell lines. Conversely, ex vivo treatment of primary AML samples with a CD40 agonist significantly decreased cell apoptosis and drug sensitivity. In Kasumi-1 AML cells, CD40 knockout (KO) significantly impaired the engraftment ability of leukemia cells and reduced the leukemia burden in NSG mice compared to wild-type mice. RNA sequencing showed that differentially expressed genes were significantly enriched in the nuclear factor-kB (NF-kB) signaling pathway in CD40-KO cells, which was confirmed through Western blotting. Untargeted metabolomic analysis revealed 179 metabolites with differential expression between WT and CD40 KO cells. Subsequent analysis revealed significant changes in the main metabolic pathways, particularly the biosynthesis of unsaturated fatty acids and lipid metabolism. A targeted metabolomics study of fatty acid metabolism demonstrated that cis-5, 8, 11, 14, 17-eicosapentaenoic acid (EPA) was markedly downregulated in CD40-KO cells compared to wild-type cells. Remarkably, EPA reversed the apoptosis and cell cycle arrest induced by CD40 deletion, simultaneously reducing the drug sensitivity of CD40-KO cells. Together, our study highlights the potential of CD40 as a target in the treatment of AML.
    DOI:  https://doi.org/10.1016/j.intimp.2025.114665
  25. Sci Rep. 2025 Apr 23. 15(1): 14217
      Cervical cancer (CC) remains a leading cause of cancer-related deaths worldwide and still requires effective interventions to improve patient outcomes. Angiopoietin-like 4 (ANGPTL4) is a multifaceted glycoprotein that plays crucial roles in lipid metabolism and tumor progression. ANGPTL4 exhibits both tumor-promoting and tumor-suppressing effects and has been proposed as a promising target for cancer therapy. This study investigated the role and potential of ANGPTL4 in enhancing therapeutic efficacy in CC using cell line models in vitro. Our analysis revealed a decreased expression of ANGPTL4 in CC samples from the GSE dataset and in the CC cell lines examined. Functional assays demonstrated that ANGPTL4 overexpression suppressed CC cell proliferation, migration, and invasion. Notably, overexpression of ANGPTL4 resulted in decreased cell viability and increased levels of apoptosis, cleaved caspase-3, and cleaved PARP under cisplatin treatment. Furthermore, these analyses were also conducted in ANGPTL4-knockdown cells, and results supporting the tumor-suppressive roles of ANGPTL4 were observed. Taken together, our study elucidates the critical role of ANGPTL4 in modulating progression and chemosensitivity of CC cells, suggesting ANGPTL4 as a potential target for CC treatment.
    Keywords:  Angiopoietin-like 4 (ANGPTL4); Apoptosis; Cervical cancer; Chemosensitivity; Cisplatin
    DOI:  https://doi.org/10.1038/s41598-025-99136-z
  26. Cell Death Dis. 2025 Apr 22. 16(1): 326
      The aggressive phenotype of prostate cancer (PCa) requires adaptation to androgen deprivation (AD) to progress into castration-resistant PCa (CRPC), including adaptation to AD-induced oxidative stress. However, our understanding of the oncogenes that maintain the redox balance during CRPC progression is limited. Here, we identified Bromodomain-containing protein 9 (BRD9) as a metabolic checkpoint for reprogramming cell metabolism to support tumor growth and impart a castration-resistant phenotype under metabolic and oxidative stress. Following oxidation, BRD9 recruited the nuclear transcription factor-Y A-subunit (NFYA) to induce glycogen phosphorylase L (PYGL) expression, which directed glucose utilization through the pentose phosphate pathway, generating NADPH, and promoting clearance of reactive oxygen species (ROS), thus maintaining redox balance. By disturbing redox homeostasis, BRD9 inhibition exerted oxidative pressure on PCa cells, sensitizing them to radiotherapy. This work identified BRD9 as a novel component in antioxidant reprogramming and indicates BRD9 targeting as a promising treatment strategy for PCa therapy.
    DOI:  https://doi.org/10.1038/s41419-025-07561-9
  27. Anticancer Drugs. 2025 Apr 23.
      The overexpression of CAV1 in many cancers is linked to chemotherapy resistance, but the exact mechanisms by which CAV1 contributes to resistance in nasopharyngeal carcinoma (NPC) are not fully known. Our research aims to elucidate the potential pathways by which CAV1 contributes to chemotherapy resistance in NPC, providing a basis for developing strategies to overcome resistance. A docetaxel-resistant NPC cell line was established, and CAV1 expression was analyzed in the cell line and the resistant variant using western blot. The sensitivity of the resistant cell line to docetaxel was assessed via cell counting kit-8, colony formation assays, and flow cytometry. Flow cytometry was used to measure lipid reactive oxygen species levels, while kits were employed to determine Fe2+ and malondialdehyde concentrations. The Ubibrowser database helped identify ubiquitination enzymes that interact with CAV1. The binding relationship between UCHL1 and CAV1 was studied using co-immunoprecipitation and immunofluorescence, which also evaluated the deubiquitination activity of UCHL1 on CAV1. CAV1 is overexpressed in NPC tissues and cells, correlating with adverse patient prognoses. In docetaxel-resistant cells, CAV1 expression is elevated compared to standard NPC cells. Silencing CAV1 increased the sensitivity of these resistant cells to docetaxel. Additionally, treatment with the ferroptosis inducer erastin could counteract the effects of CAV1 overexpression on drug resistance. UCHL1 interacted with CAV1 and inhibited its ubiquitin-mediated degradation pathway. By deubiquitinating CAV1, UCHL1 stabilizes and increases its expression, which inhibits ferroptosis and enhances the resistance of NPC cells to docetaxel.
    Keywords:  CAV1; UCHL1; docetaxel resistance; ferroptosis; nasopharyngeal carcinoma
    DOI:  https://doi.org/10.1097/CAD.0000000000001721
  28. Stem Cells. 2025 Apr 22. pii: sxaf024. [Epub ahead of print]
      CD44, a multifunctional cell surface protein, has emerged as a pivotal regulator in cancer stem cell (CSC) biology, orchestrating processes such as stemness, metabolic reprogramming, and therapeutic resistance. Recent studies have identified a critical role of CD44 in ferroptosis resistance by stabilizing SLC7A11 (xCT), a key component of the antioxidant defense system, enabling CSCs to evade oxidative stress and sustain tumorigenic potential. Additionally, CD44 regulates intracellular iron metabolism and redox balance, further supporting CSC survival and adaptation to stressful microenvironments. Therapeutic strategies targeting CD44, including ferroptosis inducers and combination therapies, have shown significant potential in preclinical and early clinical settings. Innovations such as CD44-mediated nanocarriers and metabolic inhibitors present novel opportunities to disrupt CSC-associated resistance mechanisms. Furthermore, the dynamic plasticity of CD44 isoforms governed by transcriptional, post-transcriptional, and epigenetic regulation underscores the importance of context-specific therapeutic approaches. This review highlights the multifaceted roles of CD44 in CSC biology, focusing on its contribution to ferroptosis resistance, iron metabolism, and redox regulation. Targeting CD44 offers a promising avenue for overcoming therapeutic resistance and improving the outcomes of refractory cancers. Future studies are needed to refine these strategies and enable their clinical translation.
    Keywords:  CD44; Cancer stem cell; Ferroptosis; Iron metabolism; SLC7A11
    DOI:  https://doi.org/10.1093/stmcls/sxaf024