Background: Tumor-associated neutrophils and macrophages are key components of the hepatocellular carcinoma (HCC) microenvironment. However, the interplay between them and its contribution to HCC progression remain unclear.
Methods: Bioinformatic analysis of TCGA datasets and clinical HCC samples was used to evaluate neutrophil extracellular trap (NETs) levels and macrophage polarization. Co-culture of neutrophils, macrophages, and HCC cells, along with molecular analysis and in vivo mouse models, were employed to dissect the mechanisms underlying NETs-mediated macrophage reprogramming and tumor progression.
Results: NETs were significantly elevated in HCC patients, particularly in advanced and metastatic stages, which were positively correlated with intrahepatic M2 macrophage infiltration and M2d subset-associated cytokines in blood. In vitro, NETs promoted M2d polarization in the presence of HCC cells via IL-17R/NF-κB signaling activated by IL-17 carried within NETs, which subsequently enhanced angiogenesis, migration, invasion, and epithelial-mesenchymal transition; these effects were partially reversed by IL-17R inhibition. In vivo, NETs-induced M2d polarization accelerated tumor growth, angiogenesis, and metastasis, whereas IL-17R blockade attenuated these pro-tumor effects. Moreover, M2d macrophages indirectly promoted NETs formation by upregulating HCC cell-derived S100A9 through VEGF-NF-κB signaling, establishing a positive feedback loop between neutrophils and macrophages. Furthermore, IL-17 carried by NETs (NETs-IL-17) demonstrated strong predictive value for extrahepatic metastasis in HCC, with an area under the ROC curve (AUC) of 0.89.
Conclusions: A positive feedback loop between neutrophils and macrophages via the NETs-IL-17/VEGF/S100A9 axis accelerates HCC progression and metastasis. More importantly, NETs-IL-17 exhibited potential as an alternative biomarker for predicting extrahepatic metastasis in HCC.
{"title":"Neutrophil-macrophage crosstalk via NETs-IL-17/VEGF/S100A9 axis promotes hepatocellular carcinoma progression.","authors":"Rong Wu, Rui Wu, Xuehua Kong, Xuanyi Wang, Yaqian Duan, Shiyu Cao, Shan Yu, Yuqing Zhao, Shue Li, Jingying Zhou, Liang Duan","doi":"10.1186/s13046-025-03618-x","DOIUrl":"https://doi.org/10.1186/s13046-025-03618-x","url":null,"abstract":"<p><strong>Background: </strong>Tumor-associated neutrophils and macrophages are key components of the hepatocellular carcinoma (HCC) microenvironment. However, the interplay between them and its contribution to HCC progression remain unclear.</p><p><strong>Methods: </strong>Bioinformatic analysis of TCGA datasets and clinical HCC samples was used to evaluate neutrophil extracellular trap (NETs) levels and macrophage polarization. Co-culture of neutrophils, macrophages, and HCC cells, along with molecular analysis and in vivo mouse models, were employed to dissect the mechanisms underlying NETs-mediated macrophage reprogramming and tumor progression.</p><p><strong>Results: </strong>NETs were significantly elevated in HCC patients, particularly in advanced and metastatic stages, which were positively correlated with intrahepatic M2 macrophage infiltration and M2d subset-associated cytokines in blood. In vitro, NETs promoted M2d polarization in the presence of HCC cells via IL-17R/NF-κB signaling activated by IL-17 carried within NETs, which subsequently enhanced angiogenesis, migration, invasion, and epithelial-mesenchymal transition; these effects were partially reversed by IL-17R inhibition. In vivo, NETs-induced M2d polarization accelerated tumor growth, angiogenesis, and metastasis, whereas IL-17R blockade attenuated these pro-tumor effects. Moreover, M2d macrophages indirectly promoted NETs formation by upregulating HCC cell-derived S100A9 through VEGF-NF-κB signaling, establishing a positive feedback loop between neutrophils and macrophages. Furthermore, IL-17 carried by NETs (NETs-IL-17) demonstrated strong predictive value for extrahepatic metastasis in HCC, with an area under the ROC curve (AUC) of 0.89.</p><p><strong>Conclusions: </strong>A positive feedback loop between neutrophils and macrophages via the NETs-IL-17/VEGF/S100A9 axis accelerates HCC progression and metastasis. More importantly, NETs-IL-17 exhibited potential as an alternative biomarker for predicting extrahepatic metastasis in HCC.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":" ","pages":""},"PeriodicalIF":12.8,"publicationDate":"2025-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145866088","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-29DOI: 10.1186/s13046-025-03558-6
Nicola Romanini, Ratchapong Netsrithong, Maria Themeli, Marcella Tazzari
The field of human induced pluripotent stem cell (hiPSC)-derived cell therapies is rapidly advancing, offering a promising "off-the-shelf" approach for treating both solid and hematologic malignancies. Among these, hiPSC-derived Natural Killer (NK) cell therapies have gained significant traction, with several currently in clinical trials and development. NK cell-based immunotherapy has emerged as a safe and effective strategy for patients with advanced leukemia, and ongoing research is focused on optimizing its accessibility, scalability, and efficacy. A key advantage of hiPSC-derived NK cells is their genetic susceptibility, allowing for targeted enhancements in fitness, metabolism, specificity, and cytotoxicity. This overcomes the donor-dependent variability that limits autologous and allogeneic NK cell therapies, which often struggle with expansion and functional consistency. Despite their promise, hiPSC-derived NK cells present unique manufacturing challenges, requiring precise optimization to ensure reproducibility, safety, and clinical-grade scalability. In this review, we will explore what we believe to be the most impactful genetic engineering strategies to enhance hiPSC-derived NK cell function. Additionally, we will also discuss the major hurdles challenging widespread clinical adoption, including licensing constraints, production yield, regulatory ambiguities, and the complexities of multi-step genetic engineering and safety validation. Finally, we will outline the emerging therapeutic pipelines from leading biotech companies, providing a valuable and up-to-date overview of the future landscape of hiPSC-derived NK cell therapy.
{"title":"Challenges and opportunities of human iPSC-derived NK as \"Off-the-shelf\" cellular therapies.","authors":"Nicola Romanini, Ratchapong Netsrithong, Maria Themeli, Marcella Tazzari","doi":"10.1186/s13046-025-03558-6","DOIUrl":"10.1186/s13046-025-03558-6","url":null,"abstract":"<p><p>The field of human induced pluripotent stem cell (hiPSC)-derived cell therapies is rapidly advancing, offering a promising \"off-the-shelf\" approach for treating both solid and hematologic malignancies. Among these, hiPSC-derived Natural Killer (NK) cell therapies have gained significant traction, with several currently in clinical trials and development. NK cell-based immunotherapy has emerged as a safe and effective strategy for patients with advanced leukemia, and ongoing research is focused on optimizing its accessibility, scalability, and efficacy. A key advantage of hiPSC-derived NK cells is their genetic susceptibility, allowing for targeted enhancements in fitness, metabolism, specificity, and cytotoxicity. This overcomes the donor-dependent variability that limits autologous and allogeneic NK cell therapies, which often struggle with expansion and functional consistency. Despite their promise, hiPSC-derived NK cells present unique manufacturing challenges, requiring precise optimization to ensure reproducibility, safety, and clinical-grade scalability. In this review, we will explore what we believe to be the most impactful genetic engineering strategies to enhance hiPSC-derived NK cell function. Additionally, we will also discuss the major hurdles challenging widespread clinical adoption, including licensing constraints, production yield, regulatory ambiguities, and the complexities of multi-step genetic engineering and safety validation. Finally, we will outline the emerging therapeutic pipelines from leading biotech companies, providing a valuable and up-to-date overview of the future landscape of hiPSC-derived NK cell therapy.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"327"},"PeriodicalIF":12.8,"publicationDate":"2025-12-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12746639/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145858890","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-29DOI: 10.1186/s13046-025-03587-1
Han Wu, Guoqing Zhu, Qianshu Zhu, Ji Ma, Siwei Mao, Miao Ding, Jiabei Zhu, Xiaochen Tang, Zhixuan Bian, Yuhua Shan, Song Gu, Fenyong Sun, Cizhong Jiang, Qiuhui Pan
Background: Hepatoblastoma (HB) is the most common pediatric liver malignancy with an increasing incidence. However, the functional roles of 3D chromatin organization, epigenetic regulatory factors, and transcriptional reprogramming in HB pathogenesis remain poorly understood.
Methods: Integrated multi-omics analyses of HB and matched non-tumor tissues were performed, including Hi-C, H3K27ac CUT&Tag, ATAC-seq, and RNA-seq, to construct high-resolution 3D epigenomic maps and identify genes interacting with HB-specific super-enhancers (SEs). Functional assays of identified targets were conducted in cell lines and animal models. The regulatory mechanisms of SEs and upstream transcription factors (TFs) were investigated using CRISPRi-dCas9, 3C-qPCR, ChIP-qPCR, and luciferase reporter assays.
Results: Comprehensive analysis identified TRIB2 as an HB-specific SE-associated oncogene. Functionally, TRIB2 promoted cell proliferation and accelerated tumor growth both in vitro and in vivo. Patients with high TRIB2 expression exhibited advanced PRETEXT stage and metastasis. Mechanistically, TCF3 directly bound to both the TRIB2-SE and its promoter, promoting TRIB2 overexpression. Moreover, TRIB2 conferred resistance to ferroptosis by disrupting KEAP1-mediated ubiquitination of NRF2, thereby stabilizing NRF2 protein and enhancing antioxidant responses. The TCF3-TRIB2-NRF2 axis showed significant co-expression in HB tissues, effectively distinguished HB from normal liver tissues, and was associated with poorer overall survival.
Conclusions: Our findings reveal that TCF3 and SE mediate TRIB2 overexpression to inhibit ferroptosis via the KEAP1-NRF2 pathway and drive HB pathogenesis, providing potential diagnostic and prognostic markers for HB.
{"title":"TCF3 activates super-enhancer-driven TRIB2 overexpression to suppress ferroptosis and promote hepatoblastoma proliferation.","authors":"Han Wu, Guoqing Zhu, Qianshu Zhu, Ji Ma, Siwei Mao, Miao Ding, Jiabei Zhu, Xiaochen Tang, Zhixuan Bian, Yuhua Shan, Song Gu, Fenyong Sun, Cizhong Jiang, Qiuhui Pan","doi":"10.1186/s13046-025-03587-1","DOIUrl":"10.1186/s13046-025-03587-1","url":null,"abstract":"<p><strong>Background: </strong>Hepatoblastoma (HB) is the most common pediatric liver malignancy with an increasing incidence. However, the functional roles of 3D chromatin organization, epigenetic regulatory factors, and transcriptional reprogramming in HB pathogenesis remain poorly understood.</p><p><strong>Methods: </strong>Integrated multi-omics analyses of HB and matched non-tumor tissues were performed, including Hi-C, H3K27ac CUT&Tag, ATAC-seq, and RNA-seq, to construct high-resolution 3D epigenomic maps and identify genes interacting with HB-specific super-enhancers (SEs). Functional assays of identified targets were conducted in cell lines and animal models. The regulatory mechanisms of SEs and upstream transcription factors (TFs) were investigated using CRISPRi-dCas9, 3C-qPCR, ChIP-qPCR, and luciferase reporter assays.</p><p><strong>Results: </strong>Comprehensive analysis identified TRIB2 as an HB-specific SE-associated oncogene. Functionally, TRIB2 promoted cell proliferation and accelerated tumor growth both in vitro and in vivo. Patients with high TRIB2 expression exhibited advanced PRETEXT stage and metastasis. Mechanistically, TCF3 directly bound to both the TRIB2-SE and its promoter, promoting TRIB2 overexpression. Moreover, TRIB2 conferred resistance to ferroptosis by disrupting KEAP1-mediated ubiquitination of NRF2, thereby stabilizing NRF2 protein and enhancing antioxidant responses. The TCF3-TRIB2-NRF2 axis showed significant co-expression in HB tissues, effectively distinguished HB from normal liver tissues, and was associated with poorer overall survival.</p><p><strong>Conclusions: </strong>Our findings reveal that TCF3 and SE mediate TRIB2 overexpression to inhibit ferroptosis via the KEAP1-NRF2 pathway and drive HB pathogenesis, providing potential diagnostic and prognostic markers for HB.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"329"},"PeriodicalIF":12.8,"publicationDate":"2025-12-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12750601/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145858928","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-29DOI: 10.1186/s13046-025-03572-8
Tanvi Varadkar, Zhuoxin Zora Zhou, Jiashuai Zhang, Anusua Sarkar, Zhantao Du, Srijita Chowdhury, Hwayeon Lim, Lufang Zhou, Xiaoguang Margaret Liu
Triple-negative breast cancer (TNBC) is an aggressive and chemotherapy resistant subtype with high metastatic potential and frequent recurrence after standard treatment. While poly (ADP-ribose) polymerase inhibitors (PARPi) show efficacy in TNBCs with DNA repair deficiencies, only ~ 20% of patients respond, underscoring the need for more effective therapies. Mitochondria, as central regulators of cancer cell survival, present a compelling therapeutic target. Here, we introduce a novel gene-chemotherapy combining our mitochondria-targeted luminoptogenetics technology (cmLumiOpto), which directly disrupts mitochondrial membrane potential and induces cancer cell death, with PARPi to enhance TNBC treatment outcomes. To achieve targeted delivery, we conjugated a high-affinity anti-CD276 monoclonal antibody (mAb) that selectively binds human and mouse TNBCs to an exosome-associated adeno-associated virus (mAb-Exo-AAV). In vitro studies confirmed successful transfection, internalization, and functional expression of cmLumiOpto, leading to significantly enhanced cytotoxicity when combined with PARPi. In vivo, the combination therapy achieved a 95-100% reduction in tumor burden, suppressed patient-derived xenograft growth, and inhibited metastasis in four TNBC mouse models. Post-treatment analyses confirmed mitochondrial depolarization, downregulation of DNA replication, cytokine upregulation, and immune cell infiltration in tumor. These findings highlight the potential of mitochondria-targeted gene therapy combined with chemotherapy as a powerful and innovative strategy for TNBC treatment.
{"title":"Combine mitochondrial-targeted gene therapy and chemotherapy to treat triple-negative breast cancer.","authors":"Tanvi Varadkar, Zhuoxin Zora Zhou, Jiashuai Zhang, Anusua Sarkar, Zhantao Du, Srijita Chowdhury, Hwayeon Lim, Lufang Zhou, Xiaoguang Margaret Liu","doi":"10.1186/s13046-025-03572-8","DOIUrl":"10.1186/s13046-025-03572-8","url":null,"abstract":"<p><p>Triple-negative breast cancer (TNBC) is an aggressive and chemotherapy resistant subtype with high metastatic potential and frequent recurrence after standard treatment. While poly (ADP-ribose) polymerase inhibitors (PARPi) show efficacy in TNBCs with DNA repair deficiencies, only ~ 20% of patients respond, underscoring the need for more effective therapies. Mitochondria, as central regulators of cancer cell survival, present a compelling therapeutic target. Here, we introduce a novel gene-chemotherapy combining our mitochondria-targeted luminoptogenetics technology (cmLumiOpto), which directly disrupts mitochondrial membrane potential and induces cancer cell death, with PARPi to enhance TNBC treatment outcomes. To achieve targeted delivery, we conjugated a high-affinity anti-CD276 monoclonal antibody (mAb) that selectively binds human and mouse TNBCs to an exosome-associated adeno-associated virus (mAb-Exo-AAV). In vitro studies confirmed successful transfection, internalization, and functional expression of cmLumiOpto, leading to significantly enhanced cytotoxicity when combined with PARPi. In vivo, the combination therapy achieved a 95-100% reduction in tumor burden, suppressed patient-derived xenograft growth, and inhibited metastasis in four TNBC mouse models. Post-treatment analyses confirmed mitochondrial depolarization, downregulation of DNA replication, cytokine upregulation, and immune cell infiltration in tumor. These findings highlight the potential of mitochondria-targeted gene therapy combined with chemotherapy as a powerful and innovative strategy for TNBC treatment.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"328"},"PeriodicalIF":12.8,"publicationDate":"2025-12-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12750853/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145858949","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-27DOI: 10.1186/s13046-025-03604-3
Yuanyuan Ma, Chang Guo, Xijun Wang, Xundong Wei, Jie Ma
{"title":"Correction: Impact of chemotherapeutic agents on liver microenvironment: oxaliplatin create a pro-metastatic landscape.","authors":"Yuanyuan Ma, Chang Guo, Xijun Wang, Xundong Wei, Jie Ma","doi":"10.1186/s13046-025-03604-3","DOIUrl":"10.1186/s13046-025-03604-3","url":null,"abstract":"","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"326"},"PeriodicalIF":12.8,"publicationDate":"2025-12-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12743389/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145844223","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-26DOI: 10.1186/s13046-025-03612-3
Shuang Tao, Shu-Juan Xie, Li-Ting Diao, Guo Lv, Ya-Rui Hou, Yan-Xia Hu, Wan-Yi Xu, Bin Du, Zhen-Dong Xiao
{"title":"Correction: RNA‑binding protein CCDC137 activates AKT signaling and promotes hepatocellular carcinoma through a novel non‑canonical role of DGCR8 in mRNA localization.","authors":"Shuang Tao, Shu-Juan Xie, Li-Ting Diao, Guo Lv, Ya-Rui Hou, Yan-Xia Hu, Wan-Yi Xu, Bin Du, Zhen-Dong Xiao","doi":"10.1186/s13046-025-03612-3","DOIUrl":"10.1186/s13046-025-03612-3","url":null,"abstract":"","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"325"},"PeriodicalIF":12.8,"publicationDate":"2025-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12742200/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145844160","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-26DOI: 10.1186/s13046-025-03620-3
Stefania Elena Navone, Giovanni Marfia, Laura Guarnaccia, Massimiliano Rizzaro, Giorgio Fiore, Rolando Campanella, Chiara Gaudino, Daniele Santini, Giovanni Andrea Alotta, Monica Rosa Miozzo, Emanuela Barilla, Marco Locatelli, Laura Riboni
Background: Glioblastoma is the most aggressive primary brain tumor, and, despite intensive studies, remains one of the most fatal malignancy in adult humans. Among multiple onco-promoters produced by glioblastoma cells, erythropoietin was found. However, the presence/function of Epo alternatively spliced variants in cancer remains unexplored. Here, we investigated the expression and role of Epo-variants in glioblastoma, and the therapeutic potential of their targeting through a novel monoclonal antibody (mAb).
Methods: Transcripts and protein levels of Epo-variants in a cohort of human brain tumors were evaluated by RT-PCR, ELISA, and immunohistochemistry. Monoclonal antibodies targeting Epo-Vs were prepared and functionally selected by assaying proliferation, migration, stemness, and angiogenesis in glioblastoma patient-derived cells. Antibody affinity for Epo/Epo-variant was determined by SPR. In vivo toxicity and therapeutic efficacy of the lead antibody were evaluated in GBM mouse models.
Results: We found a significant overexpression of Epo-variant transcripts in tissues and cells from GBM patients. After functional selection of newly-produced antibodies, we identified AND-C4 as the lead one for its potent anti-tumoral properties, absence of anti-erythropoietic effects and of toxicity on human brain cells. AND-C4 exhibited high affinity for the Epo-variant EV-3. We demonstrated that EV-3 was efficiently produced and secreted by glioblastoma cells, particularly by stem cells. EV-3 exerted tumorigenic, angiogenic and immunomodulatory properties, and AND-C4 was effective in antagonizing all these actions. In vivo studies in rodent glioblastoma models revealed that AND-C4 selectively bound to tumor tissue and exhibited significant efficacy on tumor growth and animal survival.
Conclusion: This study represents the first evidence on the presence, origin and pro-tumoral activity of EV-3 in human glioblastoma. Moreover, in vitro and in vivo results revealed AND-C4 as novel and promising anti-glioblastoma immunotherapeutic.
{"title":"Revealing erythropoietin variant EV-3 as novel driving force and immunotherapeutic target in human glioblastoma.","authors":"Stefania Elena Navone, Giovanni Marfia, Laura Guarnaccia, Massimiliano Rizzaro, Giorgio Fiore, Rolando Campanella, Chiara Gaudino, Daniele Santini, Giovanni Andrea Alotta, Monica Rosa Miozzo, Emanuela Barilla, Marco Locatelli, Laura Riboni","doi":"10.1186/s13046-025-03620-3","DOIUrl":"https://doi.org/10.1186/s13046-025-03620-3","url":null,"abstract":"<p><strong>Background: </strong>Glioblastoma is the most aggressive primary brain tumor, and, despite intensive studies, remains one of the most fatal malignancy in adult humans. Among multiple onco-promoters produced by glioblastoma cells, erythropoietin was found. However, the presence/function of Epo alternatively spliced variants in cancer remains unexplored. Here, we investigated the expression and role of Epo-variants in glioblastoma, and the therapeutic potential of their targeting through a novel monoclonal antibody (mAb).</p><p><strong>Methods: </strong>Transcripts and protein levels of Epo-variants in a cohort of human brain tumors were evaluated by RT-PCR, ELISA, and immunohistochemistry. Monoclonal antibodies targeting Epo-Vs were prepared and functionally selected by assaying proliferation, migration, stemness, and angiogenesis in glioblastoma patient-derived cells. Antibody affinity for Epo/Epo-variant was determined by SPR. In vivo toxicity and therapeutic efficacy of the lead antibody were evaluated in GBM mouse models.</p><p><strong>Results: </strong>We found a significant overexpression of Epo-variant transcripts in tissues and cells from GBM patients. After functional selection of newly-produced antibodies, we identified AND-C4 as the lead one for its potent anti-tumoral properties, absence of anti-erythropoietic effects and of toxicity on human brain cells. AND-C4 exhibited high affinity for the Epo-variant EV-3. We demonstrated that EV-3 was efficiently produced and secreted by glioblastoma cells, particularly by stem cells. EV-3 exerted tumorigenic, angiogenic and immunomodulatory properties, and AND-C4 was effective in antagonizing all these actions. In vivo studies in rodent glioblastoma models revealed that AND-C4 selectively bound to tumor tissue and exhibited significant efficacy on tumor growth and animal survival.</p><p><strong>Conclusion: </strong>This study represents the first evidence on the presence, origin and pro-tumoral activity of EV-3 in human glioblastoma. Moreover, in vitro and in vivo results revealed AND-C4 as novel and promising anti-glioblastoma immunotherapeutic.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":" ","pages":""},"PeriodicalIF":12.8,"publicationDate":"2025-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145835158","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-24DOI: 10.1186/s13046-025-03568-4
Zhike Zhou, Ye Hua, Jun Ma, Wenqiang Cong, Rui Zhan, Kexin Kang, Lu Wang, Hongyi Wei
Background: Triple-negative breast cancer (TNBC) is highly aggressive tumor with limited therapeutic options. Studying the molecular mechanisms underlying TNBC is necessary to address the unmet need in novel therapeutic targets. TNBC is demonstrated to have robust fatty acid (FA) metabolism activity, and recent studies proposed the linkage of FA metabolism with ferroptosis sensitivity. Hence, this study aimed to explore the targets that may regulate FA metabolism to sensitize TNBC cells to ferroptosis.
Methods: RNA-sequencing data in The Cancer Genome Atlas (TCGA) and four microarray datasets in Gene Expression Omnibus (GEO) database were analyzed to identify key target RACGAP1, followed by a series of functional experiments to explore the exact role of RACGAP1 in two TNBC cell lines (human MDA-MB-231 and mouse 4T1) and Xenograft tumor model. Dual-luciferase and chromatin immunoprecipitation (ChIP) assay was utilized to verify the binding of RACGAP1 and MAZ. RNA sequencing on 4T1 cells transfecting with sh-NC and sh-RACGAP1 was performed to validate the actions of RACGAP1.
Results: RACGAP1 was highly expressed in breast cancer, and associated with poor prognosis and ferroptosis activity. RACGAP1 silencing could inhibit tumor cells survival and promote ferroptosis, and such anti-tumor activity could be blocked by ferroptosis inhibitors. RNA-sequencing analysis suggested that RACGAP1 silencing could inhibit FA metabolism activity, which was further confirmed by metabolic analysis and the reduced level of ATP, triglyceride and FA oxidation. CPT1A overexpression reversed such changes, indicating that the regulation of RACGAP1 on FA metabolism was CPT1A-dependent. Activation of FA metabolism activity or CPT1A overexpression blocked the ferroptosis sensitivity induced by RACGAP1 silencing. Transcription factor MAZ was identified to directly up-regulate the expression of RACGAP1.
Conclusion: Inhibition of RACGAP1 sensitized TNBC cells to ferroptosis by inhibiting CPT1A-mediated FA metabolism. Targeting RACGAP1 might be feasible strategy for TNBC management.
背景:三阴性乳腺癌(TNBC)是一种高度侵袭性的肿瘤,治疗选择有限。研究TNBC的分子机制对于解决新的治疗靶点的需求是必要的。TNBC被证明具有强大的脂肪酸(FA)代谢活性,最近的研究提出FA代谢与铁下垂敏感性的联系。因此,本研究旨在探索可能调节FA代谢使TNBC细胞对铁下垂敏感的靶点。方法:分析Cancer Genome Atlas (TCGA)中的rna测序数据和Gene Expression Omnibus (GEO)数据库中的4个微阵列数据集,确定关键靶点RACGAP1,并进行一系列功能实验,探讨RACGAP1在两种TNBC细胞系(人MDA-MB-231和小鼠4T1)和Xenograft肿瘤模型中的确切作用。采用双荧光素酶和染色质免疫沉淀(ChIP)法验证RACGAP1与MAZ的结合。对转染sh-NC和sh-RACGAP1的4T1细胞进行RNA测序,验证RACGAP1的作用。结果:RACGAP1在乳腺癌中高表达,与不良预后和铁下垂活性相关。沉默RACGAP1可抑制肿瘤细胞存活,促进铁下垂,这种抗肿瘤活性可被铁下垂抑制剂阻断。rna测序分析表明,RACGAP1沉默可以抑制FA代谢活性,代谢分析和ATP、甘油三酯和FA氧化水平的降低进一步证实了这一点。CPT1A过表达逆转了这一变化,表明RACGAP1对FA代谢的调节依赖于CPT1A。激活FA代谢活性或CPT1A过表达可阻断RACGAP1沉默诱导的铁下垂敏感性。发现转录因子MAZ可直接上调RACGAP1的表达。结论:抑制RACGAP1通过抑制cpt1a介导的FA代谢而使TNBC细胞致敏。靶向RACGAP1可能是TNBC治疗的可行策略。
{"title":"Inhibition of RACGAP1 sensitizes triple-negative breast cancer cells to ferroptosis by regulating CPT1A-dependent fatty acid metabolism.","authors":"Zhike Zhou, Ye Hua, Jun Ma, Wenqiang Cong, Rui Zhan, Kexin Kang, Lu Wang, Hongyi Wei","doi":"10.1186/s13046-025-03568-4","DOIUrl":"10.1186/s13046-025-03568-4","url":null,"abstract":"<p><strong>Background: </strong>Triple-negative breast cancer (TNBC) is highly aggressive tumor with limited therapeutic options. Studying the molecular mechanisms underlying TNBC is necessary to address the unmet need in novel therapeutic targets. TNBC is demonstrated to have robust fatty acid (FA) metabolism activity, and recent studies proposed the linkage of FA metabolism with ferroptosis sensitivity. Hence, this study aimed to explore the targets that may regulate FA metabolism to sensitize TNBC cells to ferroptosis.</p><p><strong>Methods: </strong>RNA-sequencing data in The Cancer Genome Atlas (TCGA) and four microarray datasets in Gene Expression Omnibus (GEO) database were analyzed to identify key target RACGAP1, followed by a series of functional experiments to explore the exact role of RACGAP1 in two TNBC cell lines (human MDA-MB-231 and mouse 4T1) and Xenograft tumor model. Dual-luciferase and chromatin immunoprecipitation (ChIP) assay was utilized to verify the binding of RACGAP1 and MAZ. RNA sequencing on 4T1 cells transfecting with sh-NC and sh-RACGAP1 was performed to validate the actions of RACGAP1.</p><p><strong>Results: </strong>RACGAP1 was highly expressed in breast cancer, and associated with poor prognosis and ferroptosis activity. RACGAP1 silencing could inhibit tumor cells survival and promote ferroptosis, and such anti-tumor activity could be blocked by ferroptosis inhibitors. RNA-sequencing analysis suggested that RACGAP1 silencing could inhibit FA metabolism activity, which was further confirmed by metabolic analysis and the reduced level of ATP, triglyceride and FA oxidation. CPT1A overexpression reversed such changes, indicating that the regulation of RACGAP1 on FA metabolism was CPT1A-dependent. Activation of FA metabolism activity or CPT1A overexpression blocked the ferroptosis sensitivity induced by RACGAP1 silencing. Transcription factor MAZ was identified to directly up-regulate the expression of RACGAP1.</p><p><strong>Conclusion: </strong>Inhibition of RACGAP1 sensitized TNBC cells to ferroptosis by inhibiting CPT1A-mediated FA metabolism. Targeting RACGAP1 might be feasible strategy for TNBC management.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"323"},"PeriodicalIF":12.8,"publicationDate":"2025-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12729191/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145828990","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-24DOI: 10.1186/s13046-025-03585-3
Jiancheng Lv, Kai Li, Jiatong Zhou, Ruixi Yu, Qiang Lu, Ben Liu
Background: The limited response rate and substantial interindividual variability in immunotherapy outcomes remain major barriers to improving prognosis in patients with bladder cancer (BCa). As central effectors of antitumor immunity, the extent of CD8 + T cell infiltration into tumors is a key determinant of immunotherapy response. Members of the histone deacetylase (HDAC) family play critical roles in modulating tumor immune evasion and sensitivity to immunotherapy, making HDAC inhibitors of clinical interest.
Methods: A retrospective analysis was performed using data from the IMvigor210 clinical trial and follow-up data from patients with locally advanced BCa who received adjuvant immunotherapy at our center, assessing the association between HDAC1-11 expression and immunotherapy response. RNA sequencing, gene set enrichment analysis (GSEA), chromatin immunoprecipitation PCR (ChIP-PCR), co-immunoprecipitation (Co-IP), mass spectrometry, lysine site mutagenesis, RNA immunoprecipitation, and bioinformatics analysis were employed to outline the HDAC7-BTRC-SRSF7-CCL5 pathway. The immunoregulatory function of HDAC7 was evaluated using CD8 + T cell co-culture assays and tumor models in humanized NOG (HuNOG) mice. Virtual screening, MicroScale Thermophoresis (MST), and HDAC activity assays were conducted to identify potential HDAC7 specific inhibitor. The immunosensitizing effect of Pinocembrin on BCa immunotherapy was validated using a C57BL/6 mouse tumor-bearing model.
Results: Among the HDAC family members, only HDAC7 expression was significantly associated with immunotherapy response. HDAC7 was overexpressed in BCa and correlated with poorer prognosis. Functional assays demonstrated that HDAC7 suppresses CD8 + T cell infiltration, thereby reducing sensitivity to PD-1 antibody treatment. Mechanistically, HDAC7 reduced acetylation at lysine 24 of the splicing regulator SRSF7, enhancing BTRC-mediated ubiquitination and degradation of SRSF7, which promoted the processing and expression of CCL5 mRNA-a chemokine essential for CD8 + T cell recruitment. Furthermore, Pinocembrin was identified as a selective HDAC7 inhibitor that restores CD8 + T cell infiltration and improves immunotherapy efficacy in BCa.
Conclusions: HDAC7 represents a promising diagnostic and therapeutic target in BCa immunotherapy. Pinocembrin, as a specific HDAC7 inhibitor, holds potential as a combination therapy agent to improve immunotherapy response in BCa.
{"title":"Histone deacetylase HDAC7 restricts CD8 + T cell tumor infiltration and limits immunotherapy sensitivity in bladder cancer: reversal by pinocembrin.","authors":"Jiancheng Lv, Kai Li, Jiatong Zhou, Ruixi Yu, Qiang Lu, Ben Liu","doi":"10.1186/s13046-025-03585-3","DOIUrl":"10.1186/s13046-025-03585-3","url":null,"abstract":"<p><strong>Background: </strong>The limited response rate and substantial interindividual variability in immunotherapy outcomes remain major barriers to improving prognosis in patients with bladder cancer (BCa). As central effectors of antitumor immunity, the extent of CD8 + T cell infiltration into tumors is a key determinant of immunotherapy response. Members of the histone deacetylase (HDAC) family play critical roles in modulating tumor immune evasion and sensitivity to immunotherapy, making HDAC inhibitors of clinical interest.</p><p><strong>Methods: </strong>A retrospective analysis was performed using data from the IMvigor210 clinical trial and follow-up data from patients with locally advanced BCa who received adjuvant immunotherapy at our center, assessing the association between HDAC1-11 expression and immunotherapy response. RNA sequencing, gene set enrichment analysis (GSEA), chromatin immunoprecipitation PCR (ChIP-PCR), co-immunoprecipitation (Co-IP), mass spectrometry, lysine site mutagenesis, RNA immunoprecipitation, and bioinformatics analysis were employed to outline the HDAC7-BTRC-SRSF7-CCL5 pathway. The immunoregulatory function of HDAC7 was evaluated using CD8 + T cell co-culture assays and tumor models in humanized NOG (HuNOG) mice. Virtual screening, MicroScale Thermophoresis (MST), and HDAC activity assays were conducted to identify potential HDAC7 specific inhibitor. The immunosensitizing effect of Pinocembrin on BCa immunotherapy was validated using a C57BL/6 mouse tumor-bearing model.</p><p><strong>Results: </strong>Among the HDAC family members, only HDAC7 expression was significantly associated with immunotherapy response. HDAC7 was overexpressed in BCa and correlated with poorer prognosis. Functional assays demonstrated that HDAC7 suppresses CD8 + T cell infiltration, thereby reducing sensitivity to PD-1 antibody treatment. Mechanistically, HDAC7 reduced acetylation at lysine 24 of the splicing regulator SRSF7, enhancing BTRC-mediated ubiquitination and degradation of SRSF7, which promoted the processing and expression of CCL5 mRNA-a chemokine essential for CD8 + T cell recruitment. Furthermore, Pinocembrin was identified as a selective HDAC7 inhibitor that restores CD8 + T cell infiltration and improves immunotherapy efficacy in BCa.</p><p><strong>Conclusions: </strong>HDAC7 represents a promising diagnostic and therapeutic target in BCa immunotherapy. Pinocembrin, as a specific HDAC7 inhibitor, holds potential as a combination therapy agent to improve immunotherapy response in BCa.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"44 1","pages":"324"},"PeriodicalIF":12.8,"publicationDate":"2025-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12729082/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145829051","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}