首页 > 最新文献

Cancer gene therapy最新文献

英文 中文
Construction of the cancer cell continuum reveals hybrid EMT state driving lung adenocarcinoma aggression. 癌细胞连续体的构建揭示了混合EMT状态驱动肺腺癌的侵袭。
IF 5 3区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2025-11-25 DOI: 10.1038/s41417-025-00991-9
Qiuhao Qu, Yixin Ma, Chao Huang, Botao Fa, Zhenni Liu, Yuhan Li, Shigao Yang, Zhengtao Xiao

The epithelial-to-mesenchymal transition (EMT) is a pivotal cellular process integral to metastasis. Emerging evidence indicates that EMT progresses through a continuum of cellular states, yet the intricate regulatory mechanisms underpinning this transition are not fully elucidated. This study aims to reconstruct the EMT spectrum in lung adenocarcinoma (LUAD) by integrating single-cell RNA sequencing data with advanced cell trajectory inference techniques. We identified five distinct EMT states across 16,310 malignant epithelial cells derived from 10 LUAD patients, encompassing epithelial, mesenchymal, and hybrid epithelial/mesenchymal (E/M) phenotypes. Notably, the hybrid E/M states exhibited the highest metastatic potential and were correlated with a poorer prognosis for patients. By modeling the EMT regulatory network, we evaluated the influence of transcription factor (TF) activation on network stability during cellular state transitions. We identified critical TFs, including TCF7, FOXA1, FOXA2, and SMAD7, which collectively regulate pathways such as TGF-β through both intrinsic EMT-related signaling and the modulation of tumor-associated macrophages. Furthermore, we developed a risk stratification model based on EMT continuum signatures, providing a novel tool for prognostic assessment. Our findings contribute to a comprehensive understanding of EMT-driven tumor evolution and open new avenues for prognostic stratification and targeted therapies in LUAD.

上皮-间质转化(EMT)是转移的关键细胞过程。新出现的证据表明,EMT通过连续的细胞状态进行,但支持这种转变的复杂调节机制尚未完全阐明。本研究旨在通过将单细胞RNA测序数据与先进的细胞轨迹推断技术相结合,重建肺腺癌(LUAD)的EMT谱。我们从10名LUAD患者的16,310个恶性上皮细胞中鉴定出5种不同的EMT状态,包括上皮、间充质和上皮/间充质杂交(E/M)表型。值得注意的是,混合E/M状态表现出最高的转移潜力,并且与患者的预后较差相关。通过建模EMT调控网络,我们评估了转录因子(TF)激活对细胞状态转变过程中网络稳定性的影响。我们确定了关键的TGF,包括TCF7、FOXA1、FOXA2和SMAD7,它们通过内在emt相关信号和肿瘤相关巨噬细胞的调节共同调节TGF-β等途径。此外,我们开发了一种基于EMT连续谱特征的风险分层模型,为预后评估提供了一种新的工具。我们的发现有助于全面理解emt驱动的肿瘤演变,并为LUAD的预后分层和靶向治疗开辟了新的途径。
{"title":"Construction of the cancer cell continuum reveals hybrid EMT state driving lung adenocarcinoma aggression.","authors":"Qiuhao Qu, Yixin Ma, Chao Huang, Botao Fa, Zhenni Liu, Yuhan Li, Shigao Yang, Zhengtao Xiao","doi":"10.1038/s41417-025-00991-9","DOIUrl":"https://doi.org/10.1038/s41417-025-00991-9","url":null,"abstract":"<p><p>The epithelial-to-mesenchymal transition (EMT) is a pivotal cellular process integral to metastasis. Emerging evidence indicates that EMT progresses through a continuum of cellular states, yet the intricate regulatory mechanisms underpinning this transition are not fully elucidated. This study aims to reconstruct the EMT spectrum in lung adenocarcinoma (LUAD) by integrating single-cell RNA sequencing data with advanced cell trajectory inference techniques. We identified five distinct EMT states across 16,310 malignant epithelial cells derived from 10 LUAD patients, encompassing epithelial, mesenchymal, and hybrid epithelial/mesenchymal (E/M) phenotypes. Notably, the hybrid E/M states exhibited the highest metastatic potential and were correlated with a poorer prognosis for patients. By modeling the EMT regulatory network, we evaluated the influence of transcription factor (TF) activation on network stability during cellular state transitions. We identified critical TFs, including TCF7, FOXA1, FOXA2, and SMAD7, which collectively regulate pathways such as TGF-β through both intrinsic EMT-related signaling and the modulation of tumor-associated macrophages. Furthermore, we developed a risk stratification model based on EMT continuum signatures, providing a novel tool for prognostic assessment. Our findings contribute to a comprehensive understanding of EMT-driven tumor evolution and open new avenues for prognostic stratification and targeted therapies in LUAD.</p>","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":" ","pages":""},"PeriodicalIF":5.0,"publicationDate":"2025-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145602516","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
TFF3 sensitizes cervical carcinoma cells to cisplatin toxicity by binding to IGF2R TFF3通过与IGF2R结合使宫颈癌细胞对顺铂毒性增敏。
IF 5 3区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2025-11-24 DOI: 10.1038/s41417-025-00990-w
Yaxin Zhang, Liping Yan, Na An, Jiayu Geng, Wei Zhao, Bohao Feng, Yanlong Zheng, Shaojun Liang, Lei Yang
Cisplatin resistance causes ineffectiveness of cisplatin-based treatment for cervical carcinoma. The combination of cisplatin and other chemotherapeutic drugs is an available strategy to overcome this problem. However, chemotherapeutic drugs combined with cisplatin may show tissue toxicity and systemic side effects. Thus, there is a great need of seeking effective substitutes for these chemotherapeutic drugs to improve combination therapy. Here, we found that inactivating IL-6/JAK2/STAT3 signaling pathway sensitized carcinoma cells to cisplatin toxicity by increasing cisplatin accumulation, impairing DNA damage repair, and inhibiting the initiation and development of autophagy, which subsequently caused the increases in DNA damage levels and apoptosis rates in cisplatin-treated cells. We predicted that TFF3 negatively regulated transduction in the IL-6/JAK2/STAT3 pathway based on in silico analysis of the differentially expressed genes (DEGs) between highly trefoil factor 3(TFF3)-encoding mRNA-expressing carcinoma tissues and low-expressing counterparts, and experimentally determined that both ectopic expression of TFF3-encoding gene and TFF3 administration inhibited IL-6-induced STAT3 activation in carcinoma cells. Mechanistically, upon binding to IGF2R, TFF3 stabilized IGF2R by inhibiting the ubiquitin-proteasome degradation pathway to inactivate Akt and thereby STAT3. Moreover, we discovered that TFF3 administration antagonized protective effects of IL-6 stimulation against tumor-killing capacity of cisplatin. Based on these findings, we consider that TFF3 may be employed as a cisplatin sensitizer and have advantages over traditional chemotherapeutic drugs in cisplatin-based combination therapy, since it is a naturally occurring protein in cervical tissue.
顺铂耐药导致宫颈癌顺铂治疗无效。顺铂联合其他化疗药物是克服这一问题的有效策略。然而,化疗药物联合顺铂可能出现组织毒性和全身副作用。因此,迫切需要寻找这些化疗药物的有效替代品,以改善联合治疗。本研究发现,灭活IL-6/JAK2/STAT3信号通路会增加顺铂的积累,损害DNA损伤修复,抑制自噬的发生和发展,从而导致顺铂处理细胞中DNA损伤水平和凋亡率的增加,从而使癌细胞对顺铂毒性敏感。我们通过对高三叶因子3(TFF3)编码mrna表达的癌组织与低三叶因子3编码mrna表达的癌组织差异表达基因(DEGs)的计算机分析,预测TFF3负向调控IL-6/JAK2/STAT3通路的转导,并通过实验确定TFF3编码基因的异位表达和TFF3给药抑制IL-6诱导的癌细胞中STAT3的激活。在机制上,与IGF2R结合后,TFF3通过抑制泛素-蛋白酶体降解途径来稳定IGF2R,从而使Akt失活,进而使STAT3失活。此外,我们发现TFF3可以拮抗IL-6刺激对顺铂肿瘤杀伤能力的保护作用。基于这些发现,我们认为TFF3可能被用作顺铂致敏剂,并且在顺铂联合治疗中具有优于传统化疗药物的优势,因为它是宫颈组织中天然存在的蛋白质。
{"title":"TFF3 sensitizes cervical carcinoma cells to cisplatin toxicity by binding to IGF2R","authors":"Yaxin Zhang,&nbsp;Liping Yan,&nbsp;Na An,&nbsp;Jiayu Geng,&nbsp;Wei Zhao,&nbsp;Bohao Feng,&nbsp;Yanlong Zheng,&nbsp;Shaojun Liang,&nbsp;Lei Yang","doi":"10.1038/s41417-025-00990-w","DOIUrl":"10.1038/s41417-025-00990-w","url":null,"abstract":"Cisplatin resistance causes ineffectiveness of cisplatin-based treatment for cervical carcinoma. The combination of cisplatin and other chemotherapeutic drugs is an available strategy to overcome this problem. However, chemotherapeutic drugs combined with cisplatin may show tissue toxicity and systemic side effects. Thus, there is a great need of seeking effective substitutes for these chemotherapeutic drugs to improve combination therapy. Here, we found that inactivating IL-6/JAK2/STAT3 signaling pathway sensitized carcinoma cells to cisplatin toxicity by increasing cisplatin accumulation, impairing DNA damage repair, and inhibiting the initiation and development of autophagy, which subsequently caused the increases in DNA damage levels and apoptosis rates in cisplatin-treated cells. We predicted that TFF3 negatively regulated transduction in the IL-6/JAK2/STAT3 pathway based on in silico analysis of the differentially expressed genes (DEGs) between highly trefoil factor 3(TFF3)-encoding mRNA-expressing carcinoma tissues and low-expressing counterparts, and experimentally determined that both ectopic expression of TFF3-encoding gene and TFF3 administration inhibited IL-6-induced STAT3 activation in carcinoma cells. Mechanistically, upon binding to IGF2R, TFF3 stabilized IGF2R by inhibiting the ubiquitin-proteasome degradation pathway to inactivate Akt and thereby STAT3. Moreover, we discovered that TFF3 administration antagonized protective effects of IL-6 stimulation against tumor-killing capacity of cisplatin. Based on these findings, we consider that TFF3 may be employed as a cisplatin sensitizer and have advantages over traditional chemotherapeutic drugs in cisplatin-based combination therapy, since it is a naturally occurring protein in cervical tissue.","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":"33 1","pages":"155-168"},"PeriodicalIF":5.0,"publicationDate":"2025-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145595723","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
mTORC1/2 inhibition induces tumor regression in CDK4/6 inhibitor-insensitive acral melanoma. mTORC1/2抑制诱导CDK4/6抑制剂不敏感肢端黑色素瘤的肿瘤消退。
IF 5 3区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2025-11-24 DOI: 10.1038/s41417-025-00987-5
Jeremy A Bravo Narula, Kasturee Jagirdar, Sumaiya Begum, Kuai Yu, Meihan Wei, Bailey Roberson, Marie Portuallo, Limin An, Steffanus Hallis, Viviana A Smart, Manoel O Moraes Junior, Qin Liu, Hongkai Ji, Vito W Rebecca

Acral melanoma (AM), a rare and aggressive subtype with 5-year survival rates below 16%, exhibits limited response to CDK4/6 inhibitors (CDK4i/6i) despite frequent pathway alterations. Here, we identify AKT-mTOR signaling as a critical escape mechanism triggered by CDK4/6 inhibition. Using a genetically diverse panel of AM cell lines, we demonstrate that CDK4i/6i induces rapid hyperactivation of AKT (pS473) and mTORC1 (pS6 S240/244) alongside Rb dephosphorylation. Interestingly, CDK4i/6i disrupts the cytoplasmic interaction of Rb with the mTORC2 subunit Sin1, suggesting the loss of the Rb-Sin1 protein-protein interaction may lead to mTORC2-mediated AKT hyperactivation following CDK4i/6i. Pharmacological inhibition of the AKT-mTOR axis significantly increases CDK4i/6i efficacy, as seen in the ability to reduce clonogenic survival and the ability to increase annexin+ cytotoxicity relative to single-agent CDK4i/6i, AKTi, or mTORi activity alone. These findings provide preclinical rationale for co-targeting CDK4/6 and mTORC1/2 to improve AM outcomes.

肢端黑色素瘤(AM)是一种罕见的侵袭性亚型,5年生存率低于16%,尽管经常发生通路改变,但对CDK4/6抑制剂(CDK4i/6i)的反应有限。在这里,我们确定AKT-mTOR信号是CDK4/6抑制触发的关键逃逸机制。通过对AM细胞系的遗传多样性研究,我们发现CDK4i/6i可诱导AKT (pS473)和mTORC1 (pS6 S240/244)的快速过度激活,同时Rb去磷酸化。有趣的是,CDK4i/6i破坏了Rb与mTORC2亚基Sin1的胞质相互作用,这表明Rb-Sin1蛋白-蛋白相互作用的缺失可能导致CDK4i/6i之后mTORC2介导的AKT过度激活。药理抑制AKT-mTOR轴可显著提高CDK4i/6i的疗效,与单药CDK4i/6i、AKTi或mTORi活性相比,可以降低克隆存活和增加膜联蛋白+细胞毒性。这些发现为联合靶向CDK4/6和mTORC1/2以改善AM预后提供了临床前理论依据。
{"title":"mTORC1/2 inhibition induces tumor regression in CDK4/6 inhibitor-insensitive acral melanoma.","authors":"Jeremy A Bravo Narula, Kasturee Jagirdar, Sumaiya Begum, Kuai Yu, Meihan Wei, Bailey Roberson, Marie Portuallo, Limin An, Steffanus Hallis, Viviana A Smart, Manoel O Moraes Junior, Qin Liu, Hongkai Ji, Vito W Rebecca","doi":"10.1038/s41417-025-00987-5","DOIUrl":"https://doi.org/10.1038/s41417-025-00987-5","url":null,"abstract":"<p><p>Acral melanoma (AM), a rare and aggressive subtype with 5-year survival rates below 16%, exhibits limited response to CDK4/6 inhibitors (CDK4i/6i) despite frequent pathway alterations. Here, we identify AKT-mTOR signaling as a critical escape mechanism triggered by CDK4/6 inhibition. Using a genetically diverse panel of AM cell lines, we demonstrate that CDK4i/6i induces rapid hyperactivation of AKT (pS473) and mTORC1 (pS6 S240/244) alongside Rb dephosphorylation. Interestingly, CDK4i/6i disrupts the cytoplasmic interaction of Rb with the mTORC2 subunit Sin1, suggesting the loss of the Rb-Sin1 protein-protein interaction may lead to mTORC2-mediated AKT hyperactivation following CDK4i/6i. Pharmacological inhibition of the AKT-mTOR axis significantly increases CDK4i/6i efficacy, as seen in the ability to reduce clonogenic survival and the ability to increase annexin<sup>+</sup> cytotoxicity relative to single-agent CDK4i/6i, AKTi, or mTORi activity alone. These findings provide preclinical rationale for co-targeting CDK4/6 and mTORC1/2 to improve AM outcomes.</p>","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":" ","pages":""},"PeriodicalIF":5.0,"publicationDate":"2025-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145595734","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeted suppression of tumor growth by CREPT promoter-driven diphtheria toxin fragment A 爬行启动子驱动白喉毒素片段A靶向抑制肿瘤生长。
IF 5 3区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2025-11-22 DOI: 10.1038/s41417-025-00961-1
Jingya Li, Mengdi Li, Wenchen Wang, Guancheng Jiang, Yinyin Wang, Jianqiu Sheng, Zhijie Chang, Jian Sheng, Mingyang Li, Fangli Ren
Despite significant advancements in diagnosis and treatment, cancer remains a leading cause of mortality globally. Cancer gene therapy has emerged as a promising strategy, with numerous clinical trials demonstrating its efficacy in targeting tumor cells, vasculature, and immune components. However, precise and selective gene expression regulation remains a challenge. In this study, we demonstrated that the CREPT (cell-cycle related and expression-elevated protein in tumor) promoter holds great potential for targeted cancer gene therapy. CREPT, a tumor-promoting protein as a positive regulator of gene transcription, is highly expressed across various cancer types while exhibiting minimal expression in normal tissues. The CREPT promoter mediated robust and tumor-selective transgene expression in vivo following both local and systemic administration, with only minimal off-target expression detected in blood and none in normal organs. Leveraging this specificity, we engineered an adenovirus encoding diphtheria toxin fragment A under CREPT promoter regulation. This construct was selectively expressed and inhibited protein synthesis, leading cancer cell death in vitro and in vivo. These findings demonstrate that the CREPT promoter may serve as a useful regulatory element for the development of targeted cancer gene therapy.
尽管在诊断和治疗方面取得了重大进展,但癌症仍然是全球死亡的主要原因。癌症基因治疗已经成为一种很有前途的策略,大量的临床试验证明了它在靶向肿瘤细胞、脉管系统和免疫成分方面的有效性。然而,精确和选择性的基因表达调控仍然是一个挑战。在这项研究中,我们证明了肿瘤细胞周期相关和表达升高蛋白(cellular -cycle related and expression-elevated protein In tumor)启动子在靶向癌症基因治疗中具有很大的潜力。蹑手蹑脚是一种肿瘤促进蛋白,作为基因转录的正调节因子,在各种癌症类型中高表达,而在正常组织中表达很少。在局部和全身给药后,悄悄启动子介导了体内强大的肿瘤选择性转基因表达,仅在血液中检测到少量脱靶表达,在正常器官中未检测到脱靶表达。利用这种特异性,我们设计了一种在爬行启动子调控下编码白喉毒素片段A的腺病毒。该构建体在体外和体内选择性表达并抑制蛋白质合成,导致癌细胞死亡。这些发现表明,匍匐启动子可能作为一个有用的调控元件,开发靶向癌症基因治疗。
{"title":"Targeted suppression of tumor growth by CREPT promoter-driven diphtheria toxin fragment A","authors":"Jingya Li,&nbsp;Mengdi Li,&nbsp;Wenchen Wang,&nbsp;Guancheng Jiang,&nbsp;Yinyin Wang,&nbsp;Jianqiu Sheng,&nbsp;Zhijie Chang,&nbsp;Jian Sheng,&nbsp;Mingyang Li,&nbsp;Fangli Ren","doi":"10.1038/s41417-025-00961-1","DOIUrl":"10.1038/s41417-025-00961-1","url":null,"abstract":"Despite significant advancements in diagnosis and treatment, cancer remains a leading cause of mortality globally. Cancer gene therapy has emerged as a promising strategy, with numerous clinical trials demonstrating its efficacy in targeting tumor cells, vasculature, and immune components. However, precise and selective gene expression regulation remains a challenge. In this study, we demonstrated that the CREPT (cell-cycle related and expression-elevated protein in tumor) promoter holds great potential for targeted cancer gene therapy. CREPT, a tumor-promoting protein as a positive regulator of gene transcription, is highly expressed across various cancer types while exhibiting minimal expression in normal tissues. The CREPT promoter mediated robust and tumor-selective transgene expression in vivo following both local and systemic administration, with only minimal off-target expression detected in blood and none in normal organs. Leveraging this specificity, we engineered an adenovirus encoding diphtheria toxin fragment A under CREPT promoter regulation. This construct was selectively expressed and inhibited protein synthesis, leading cancer cell death in vitro and in vivo. These findings demonstrate that the CREPT promoter may serve as a useful regulatory element for the development of targeted cancer gene therapy.","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":"33 1","pages":"145-154"},"PeriodicalIF":5.0,"publicationDate":"2025-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145581965","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PufA drives cholangiocarcinoma progression via NFκB signaling activation: a novel therapeutic target PufA通过NFκB信号激活驱动胆管癌进展:一个新的治疗靶点。
IF 5 3区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2025-11-22 DOI: 10.1038/s41417-025-00968-8
Chiao-Ping Chen, Tsai-Hsien Hung, Yenlin Huang, Yi-Ru Pan, Chun-Nan Yeh, Wen-Kuan Huang, Yu-Tien Hsiao, Chih-Hong Lo, Alice L. Yu, John Yu, Chiao-En Wu
Cholangiocarcinoma (CCA) exhibits poor prognosis despite recent advances in targeted therapy and immunotherapy. Therefore, uncovering novel mechanisms of CCA tumorigenesis and identifying effective targeted therapies are critical for improving treatment outcomes in advanced cases. PufA, a newly identified member of the Puf-family RNA-binding proteins and a biogenesis factor, is overexpressed in CCA and correlates with poor survival outcomes. Functional studies demonstrate that PufA silencing in CCA cell lines significantly reduces cell viability, induces apoptosis, and suppresses migration by inhibiting epithelial-mesenchymal transition (EMT). Additionally, PufA knockdown impairs sphere formation in vitro and inhibits tumor growth in xenograft mouse models. RNA sequencing analysis reveals significant downregulation of RELA encodes protein of p65, which encodes the p65 protein, a critical subunit for NFκB signaling activation, suggesting a connection between PufA and the NFκB signaling pathway. Mechanistic investigations confirm that PufA promotes IκBα degradation, thereby activating the NFκB signaling and regulating downstream cytokine expression, including IL1A. Conversely, PufA overexpression enhances IκBα degradation following TNFα stimulation in CCA cells, supporting the correlation between PufA and the NFκB signaling pathway. These findings establish PufA as a pivotal regulator of CCA progression and highlight the PufA-NFκB axis as a potential therapeutic target. This study provides critical insights into the molecular drivers of CCA and lays a foundation for developing novel treatments to enhance patient outcomes.
尽管最近在靶向治疗和免疫治疗方面取得了进展,但胆管癌(CCA)的预后较差。因此,揭示CCA肿瘤发生的新机制和确定有效的靶向治疗对于改善晚期病例的治疗效果至关重要。PufA是puf家族rna结合蛋白的新成员,也是一种生物发生因子,在CCA中过度表达,与较差的生存结果相关。功能研究表明,在CCA细胞系中,PufA沉默可显著降低细胞活力,诱导细胞凋亡,并通过抑制上皮-间质转化(epithelial-mesenchymal transition, EMT)抑制迁移。此外,PufA敲低可损害体外球体形成并抑制异种移植小鼠模型中的肿瘤生长。RNA测序分析显示,RELA编码p65蛋白显著下调,p65蛋白编码p65蛋白,p65蛋白是NFκB信号通路激活的关键亚基,这表明PufA与NFκB信号通路之间存在联系。机制研究证实,PufA促进i - κ b α降解,从而激活nf - κ b信号传导并调节下游细胞因子的表达,包括IL1A。相反,在CCA细胞中,PufA过表达增强TNFα刺激后i - κ b α的降解,支持PufA与nf - κ b信号通路之间的相关性。这些发现表明PufA是CCA进展的关键调节因子,并强调PufA- nfκ b轴是一个潜在的治疗靶点。这项研究为CCA的分子驱动因素提供了重要的见解,并为开发新的治疗方法奠定了基础,以提高患者的预后。
{"title":"PufA drives cholangiocarcinoma progression via NFκB signaling activation: a novel therapeutic target","authors":"Chiao-Ping Chen,&nbsp;Tsai-Hsien Hung,&nbsp;Yenlin Huang,&nbsp;Yi-Ru Pan,&nbsp;Chun-Nan Yeh,&nbsp;Wen-Kuan Huang,&nbsp;Yu-Tien Hsiao,&nbsp;Chih-Hong Lo,&nbsp;Alice L. Yu,&nbsp;John Yu,&nbsp;Chiao-En Wu","doi":"10.1038/s41417-025-00968-8","DOIUrl":"10.1038/s41417-025-00968-8","url":null,"abstract":"Cholangiocarcinoma (CCA) exhibits poor prognosis despite recent advances in targeted therapy and immunotherapy. Therefore, uncovering novel mechanisms of CCA tumorigenesis and identifying effective targeted therapies are critical for improving treatment outcomes in advanced cases. PufA, a newly identified member of the Puf-family RNA-binding proteins and a biogenesis factor, is overexpressed in CCA and correlates with poor survival outcomes. Functional studies demonstrate that PufA silencing in CCA cell lines significantly reduces cell viability, induces apoptosis, and suppresses migration by inhibiting epithelial-mesenchymal transition (EMT). Additionally, PufA knockdown impairs sphere formation in vitro and inhibits tumor growth in xenograft mouse models. RNA sequencing analysis reveals significant downregulation of RELA encodes protein of p65, which encodes the p65 protein, a critical subunit for NFκB signaling activation, suggesting a connection between PufA and the NFκB signaling pathway. Mechanistic investigations confirm that PufA promotes IκBα degradation, thereby activating the NFκB signaling and regulating downstream cytokine expression, including IL1A. Conversely, PufA overexpression enhances IκBα degradation following TNFα stimulation in CCA cells, supporting the correlation between PufA and the NFκB signaling pathway. These findings establish PufA as a pivotal regulator of CCA progression and highlight the PufA-NFκB axis as a potential therapeutic target. This study provides critical insights into the molecular drivers of CCA and lays a foundation for developing novel treatments to enhance patient outcomes.","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":"33 1","pages":"132-144"},"PeriodicalIF":5.0,"publicationDate":"2025-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145581984","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Meta single-cell atlas and xQTL post-GWAS analysis revealed the pathogenic features of thyroid cancer for target therapy: A multi-omics study Meta单细胞图谱和xQTL gwas后分析揭示了甲状腺癌靶向治疗的致病特征:一项多组学研究。
IF 5 3区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2025-11-17 DOI: 10.1038/s41417-025-00988-4
Cong Zhang, Yu Wang, Biao Xie
Thyroid cancer (TC) is an endocrine malignancy characterized by metabolic abnormalities, with its incidence continually on the rise. Understanding the pathogenesis of this cancer would help develop better diagnostic and therapeutic methods. We aimed to integrate single-cell transcriptomics, bulk transcriptomics, and GWAS data to identify causal associations with thyroid cancer at the gene level. We intended to utilize single-cell atlases to identify malignant cells and their characteristics, and employed SMR to search for genetic loci causally associated with thyroid cancer. We validated the expression differences of the genes at the single-cell level and bulk level, as well as through immunohistochemistry experimental results. We investigated the tumor immune microenvironment of patients, attempting to find immune subgroups with differential proportions. Based on these subgroups, we conducted multi-machine learning modeling to predict the likelihood of disease and developed a corresponding interactive web application. HMGA2, SDCCAG8, DLG5, MT1E, RABL2B, RERE, and NDUFA12 all demonstrated to varying degrees their roles in promoting or inhibiting the occurrence and development of thyroid cancer, with HMGA2 showing consistency across all analyses. We also identified some immune subtypes significantly associated with TC and chose markers of T_cell_C8_STMN1 to construct patient diagnostic models. Through various combinations of machine learning feature selection and model construction, we ultimately built 178 diagnostic models, with the combination of glmBoost+RF having the best diagnostic performance (Average AUPR: 0.9915). The predictive web pages ( https://zclab-cnp.shinyapps.io/TC-WEB/ ) can provide convenience and reference for clinical personnel.
甲状腺癌(TC)是一种以代谢异常为特征的内分泌恶性肿瘤,其发病率不断上升。了解这种癌症的发病机制将有助于开发更好的诊断和治疗方法。我们的目标是整合单细胞转录组学、整体转录组学和GWAS数据,在基因水平上确定与甲状腺癌的因果关系。我们打算利用单细胞图谱来鉴定恶性细胞及其特征,并利用SMR来寻找与甲状腺癌因果相关的遗传位点。我们在单细胞水平和体水平以及免疫组化实验结果验证了基因的表达差异。我们研究了患者的肿瘤免疫微环境,试图找到不同比例的免疫亚群。基于这些分组,我们进行了多机器学习建模来预测疾病的可能性,并开发了相应的交互式web应用程序。HMGA2、SDCCAG8、DLG5、MT1E、RABL2B、RERE、NDUFA12均不同程度地显示其促进或抑制甲状腺癌发生发展的作用,HMGA2在所有分析中均表现出一致性。我们还发现了一些与TC显著相关的免疫亚型,并选择了T_cell_C8_STMN1标记物来构建患者诊断模型。通过机器学习特征选择和模型构建的各种组合,我们最终构建了178个诊断模型,其中glmBoost+RF组合的诊断性能最好(平均AUPR: 0.9915)。预测网页(https://zclab-cnp.shinyapps.io/TC-WEB/)可以为临床人员提供方便和参考。
{"title":"Meta single-cell atlas and xQTL post-GWAS analysis revealed the pathogenic features of thyroid cancer for target therapy: A multi-omics study","authors":"Cong Zhang,&nbsp;Yu Wang,&nbsp;Biao Xie","doi":"10.1038/s41417-025-00988-4","DOIUrl":"10.1038/s41417-025-00988-4","url":null,"abstract":"Thyroid cancer (TC) is an endocrine malignancy characterized by metabolic abnormalities, with its incidence continually on the rise. Understanding the pathogenesis of this cancer would help develop better diagnostic and therapeutic methods. We aimed to integrate single-cell transcriptomics, bulk transcriptomics, and GWAS data to identify causal associations with thyroid cancer at the gene level. We intended to utilize single-cell atlases to identify malignant cells and their characteristics, and employed SMR to search for genetic loci causally associated with thyroid cancer. We validated the expression differences of the genes at the single-cell level and bulk level, as well as through immunohistochemistry experimental results. We investigated the tumor immune microenvironment of patients, attempting to find immune subgroups with differential proportions. Based on these subgroups, we conducted multi-machine learning modeling to predict the likelihood of disease and developed a corresponding interactive web application. HMGA2, SDCCAG8, DLG5, MT1E, RABL2B, RERE, and NDUFA12 all demonstrated to varying degrees their roles in promoting or inhibiting the occurrence and development of thyroid cancer, with HMGA2 showing consistency across all analyses. We also identified some immune subtypes significantly associated with TC and chose markers of T_cell_C8_STMN1 to construct patient diagnostic models. Through various combinations of machine learning feature selection and model construction, we ultimately built 178 diagnostic models, with the combination of glmBoost+RF having the best diagnostic performance (Average AUPR: 0.9915). The predictive web pages ( https://zclab-cnp.shinyapps.io/TC-WEB/ ) can provide convenience and reference for clinical personnel.","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":"33 1","pages":"116-131"},"PeriodicalIF":5.0,"publicationDate":"2025-11-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145538927","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
From resistance to sensitivity: the impact of FADD and lncRNA PPFIA1-AS1 on cisplatin treatment in LUSC 从耐药到敏感:FADD和lncRNA PPFIA1-AS1对LUSC顺铂治疗的影响
IF 5 3区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2025-11-14 DOI: 10.1038/s41417-025-00986-6
Xiaoyao Chang, Dangran Li, Yuanhao Tan, Fangfang Cai, Shuilian Fu, Hongqin Zhuang, Zi-Chun Hua
Cisplatin resistance remains a significant challenge in treating lung squamous cell carcinoma (LUSC). The role of FADD in this resistance requires further investigation. Our study revealed that FADD is overexpressed in LUSC patients, correlating with lower survival rates. We also discovered that long-term cisplatin-resistant LUSC cell lines (LUSC-CR) had elevated FADD protein levels, and reducing FADD restored their cisplatin sensitivity. At the same time, LUSC-CR cells resisted cisplatin-induced DNA damage and had enhanced DNA repair, linked to P53’s negative regulation of FADD. Additionally, knockdown of the long non-coding RNA (lncRNA) PPFIA1-AS1 can potentiate drug resistance in LUSC cells by decelerating FADD protein turnover and elevating FADD protein levels. In essence, this study elucidated novel mechanisms underlying cisplatin resistance in LUSC, wherein the PPFIA1-AS1/FADD axis regulates DNA damage and repair. Consequently, targeting the PPFIA1-AS1/FADD axis may present a promising avenue for overcoming cisplatin resistance and enhancing the prognosis of LUSC patients.
顺铂耐药仍然是治疗肺鳞状细胞癌(LUSC)的重大挑战。FADD在这种耐药性中的作用有待进一步研究。我们的研究表明,FADD在LUSC患者中过度表达,与较低的生存率相关。我们还发现,长期顺铂耐药的LUSC细胞系(LUSC- cr) FADD蛋白水平升高,FADD的降低恢复了它们对顺铂的敏感性。同时,LUSC-CR细胞抵抗顺铂诱导的DNA损伤,DNA修复增强,这与P53对FADD的负调控有关。此外,长链非编码RNA (lncRNA) PPFIA1-AS1的敲低可以通过减缓FADD蛋白的周转和提高FADD蛋白水平来增强LUSC细胞的耐药性。本质上,本研究阐明了LUSC顺铂耐药的新机制,其中PPFIA1-AS1/FADD轴调节DNA损伤和修复。因此,靶向PPFIA1-AS1/FADD轴可能为克服顺铂耐药和改善LUSC患者预后提供了一条有希望的途径。
{"title":"From resistance to sensitivity: the impact of FADD and lncRNA PPFIA1-AS1 on cisplatin treatment in LUSC","authors":"Xiaoyao Chang,&nbsp;Dangran Li,&nbsp;Yuanhao Tan,&nbsp;Fangfang Cai,&nbsp;Shuilian Fu,&nbsp;Hongqin Zhuang,&nbsp;Zi-Chun Hua","doi":"10.1038/s41417-025-00986-6","DOIUrl":"10.1038/s41417-025-00986-6","url":null,"abstract":"Cisplatin resistance remains a significant challenge in treating lung squamous cell carcinoma (LUSC). The role of FADD in this resistance requires further investigation. Our study revealed that FADD is overexpressed in LUSC patients, correlating with lower survival rates. We also discovered that long-term cisplatin-resistant LUSC cell lines (LUSC-CR) had elevated FADD protein levels, and reducing FADD restored their cisplatin sensitivity. At the same time, LUSC-CR cells resisted cisplatin-induced DNA damage and had enhanced DNA repair, linked to P53’s negative regulation of FADD. Additionally, knockdown of the long non-coding RNA (lncRNA) PPFIA1-AS1 can potentiate drug resistance in LUSC cells by decelerating FADD protein turnover and elevating FADD protein levels. In essence, this study elucidated novel mechanisms underlying cisplatin resistance in LUSC, wherein the PPFIA1-AS1/FADD axis regulates DNA damage and repair. Consequently, targeting the PPFIA1-AS1/FADD axis may present a promising avenue for overcoming cisplatin resistance and enhancing the prognosis of LUSC patients.","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":"33 1","pages":"102-115"},"PeriodicalIF":5.0,"publicationDate":"2025-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145523051","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Development and preclinical evaluation of ovNDV-28: a chimeric Newcastle disease virus expressing human IL-2 for cancer therapy 表达人IL-2的嵌合新城疫病毒ovNDV-28的开发和临床前评价
IF 5 3区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2025-11-04 DOI: 10.1038/s41417-025-00981-x
Tianyan Liu, Zhihang Liu, Dan Yu, Shan Jiang, Chengkai Yin, Yu Zhang, Yating Zhang, Hongna Chen, Chenfeng Zhang, Xu Li, Jiarui Yang, Shishi Liu, Zhenzhong Wang, Deshan Li
Newcastle disease virus (NDV) is a promising oncolytic virus, yet requires further optimization. In this study, we engineered an F-gene-chimeric NDV expressing human Interleukin 2 (hIL-2) to enhance the oncolytic efficacy of the NDV Clone30 strain. This recombinant virus, designated ovNDV-28, was then produced in suspension-cultured HEK293 cells. The therapeutic potential of ovNDV-28 was evaluated across multiple cancer cell lines, as well as in the HuH-7 xenograft and B16-F0 syngeneic models. Both in vitro and in vivo results demonstrated that ovNDV-28 significantly improved tumor growth suppression compared to the wild-type NDV. Flow cytometry revealed notable increases in tumor-infiltrating CD3⁺CD4⁺ T cells, CD3⁺CD8⁺ T cells, and CD3⁻CD49b⁺ cells, along with elevated expression levels of IFN-γ, TNF-α, perforin, and Granzyme B within tumor tissue. Comprehensive toxicological assessments conducted on B16-F0 tumor-bearing mice involved intratumoral administration of ovNDV-28 at doses of 1.12 × 10⁶ or 1.46 × 10⁷ PFU/mouse every other day for 14 days. No ovNDV-28-related biochemical, hematological, or histopathological abnormalities were observed. The virus was detected in tumor tissue, mesenteric lymph nodes, abdominal adipose tissue, brain, and biceps femoris, without evidence of blood circulation or viral shedding. This study systematically demonstrates the efficacy, safety, and pharmacokinetics of ovNDV-28, supporting its potential for clinical translation.
新城疫病毒(NDV)是一种很有前途的溶瘤病毒,但需要进一步优化。在这项研究中,我们设计了一种表达人白细胞介素2 (il -2)的f基因嵌合NDV,以提高NDV Clone30株的溶瘤效果。该重组病毒命名为ovNDV-28,然后在悬浮培养的HEK293细胞中产生。ovNDV-28的治疗潜力在多个癌细胞系以及HuH-7异种移植和B16-F0同基因模型中进行了评估。体外和体内实验结果均表明,与野生型NDV相比,ovNDV-28显著改善了肿瘤生长抑制。流式细胞术显示,肿瘤浸润性CD3 + CD4 + T细胞、CD3 + CD8 + T细胞和CD3⁻CD49b +细胞明显增加,肿瘤组织内IFN-γ、TNF-α、穿孔素和颗粒酶B的表达水平升高。对B16-F0荷瘤小鼠进行综合毒理学评估,每隔一天给药1.12 × 10⁶或1.46 × 10⁷PFU/小鼠,持续14天。未观察到ovndv -28相关的生化、血液学或组织病理学异常。在肿瘤组织、肠系膜淋巴结、腹部脂肪组织、大脑和股二头肌中检测到病毒,没有血液循环或病毒脱落的证据。本研究系统地证明了ovNDV-28的有效性、安全性和药代动力学,支持其临床转化的潜力。
{"title":"Development and preclinical evaluation of ovNDV-28: a chimeric Newcastle disease virus expressing human IL-2 for cancer therapy","authors":"Tianyan Liu,&nbsp;Zhihang Liu,&nbsp;Dan Yu,&nbsp;Shan Jiang,&nbsp;Chengkai Yin,&nbsp;Yu Zhang,&nbsp;Yating Zhang,&nbsp;Hongna Chen,&nbsp;Chenfeng Zhang,&nbsp;Xu Li,&nbsp;Jiarui Yang,&nbsp;Shishi Liu,&nbsp;Zhenzhong Wang,&nbsp;Deshan Li","doi":"10.1038/s41417-025-00981-x","DOIUrl":"10.1038/s41417-025-00981-x","url":null,"abstract":"Newcastle disease virus (NDV) is a promising oncolytic virus, yet requires further optimization. In this study, we engineered an F-gene-chimeric NDV expressing human Interleukin 2 (hIL-2) to enhance the oncolytic efficacy of the NDV Clone30 strain. This recombinant virus, designated ovNDV-28, was then produced in suspension-cultured HEK293 cells. The therapeutic potential of ovNDV-28 was evaluated across multiple cancer cell lines, as well as in the HuH-7 xenograft and B16-F0 syngeneic models. Both in vitro and in vivo results demonstrated that ovNDV-28 significantly improved tumor growth suppression compared to the wild-type NDV. Flow cytometry revealed notable increases in tumor-infiltrating CD3⁺CD4⁺ T cells, CD3⁺CD8⁺ T cells, and CD3⁻CD49b⁺ cells, along with elevated expression levels of IFN-γ, TNF-α, perforin, and Granzyme B within tumor tissue. Comprehensive toxicological assessments conducted on B16-F0 tumor-bearing mice involved intratumoral administration of ovNDV-28 at doses of 1.12 × 10⁶ or 1.46 × 10⁷ PFU/mouse every other day for 14 days. No ovNDV-28-related biochemical, hematological, or histopathological abnormalities were observed. The virus was detected in tumor tissue, mesenteric lymph nodes, abdominal adipose tissue, brain, and biceps femoris, without evidence of blood circulation or viral shedding. This study systematically demonstrates the efficacy, safety, and pharmacokinetics of ovNDV-28, supporting its potential for clinical translation.","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":"33 1","pages":"85-101"},"PeriodicalIF":5.0,"publicationDate":"2025-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145437363","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Correction: RUNX1A isoform is overexpressed in acute myeloid leukemia and is associated with FLT3 internal tandem duplications 纠正:RUNX1A亚型在急性髓性白血病中过表达,并与FLT3内部串联重复有关。
IF 5 3区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2025-11-03 DOI: 10.1038/s41417-025-00982-w
Cosimo Cumbo, Francesco Tarantini, Elisa Parciante, Luisa Anelli, Antonella Zagaria, Giuseppina Biondi, Mariano Francesco Caratozzolo, Paola Orsini, Nicoletta Coccaro, Giuseppina Tota, Immacolata Redavid, Maria Rosa Conserva, Angela Minervini, Crescenzio Francesco Minervini, Vito Pier Gagliardi, Mario Delia, Flaviana Marzano, Claudia Telegrafo, Sharon Natasha Cox, Annalisa Natalicchio, Bachir Balech, Apollonia Tullo, Mattia Gentile, Francesco Giorgino, Giorgina Specchia, Pellegrino Musto, Francesco Albano
{"title":"Correction: RUNX1A isoform is overexpressed in acute myeloid leukemia and is associated with FLT3 internal tandem duplications","authors":"Cosimo Cumbo,&nbsp;Francesco Tarantini,&nbsp;Elisa Parciante,&nbsp;Luisa Anelli,&nbsp;Antonella Zagaria,&nbsp;Giuseppina Biondi,&nbsp;Mariano Francesco Caratozzolo,&nbsp;Paola Orsini,&nbsp;Nicoletta Coccaro,&nbsp;Giuseppina Tota,&nbsp;Immacolata Redavid,&nbsp;Maria Rosa Conserva,&nbsp;Angela Minervini,&nbsp;Crescenzio Francesco Minervini,&nbsp;Vito Pier Gagliardi,&nbsp;Mario Delia,&nbsp;Flaviana Marzano,&nbsp;Claudia Telegrafo,&nbsp;Sharon Natasha Cox,&nbsp;Annalisa Natalicchio,&nbsp;Bachir Balech,&nbsp;Apollonia Tullo,&nbsp;Mattia Gentile,&nbsp;Francesco Giorgino,&nbsp;Giorgina Specchia,&nbsp;Pellegrino Musto,&nbsp;Francesco Albano","doi":"10.1038/s41417-025-00982-w","DOIUrl":"10.1038/s41417-025-00982-w","url":null,"abstract":"","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":"33 1","pages":"169-169"},"PeriodicalIF":5.0,"publicationDate":"2025-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.comhttps://www.nature.com/articles/s41417-025-00982-w.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145437423","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Bacterial nanoparticles as a potent and safe alternative to BCG for bladder cancer immunotherapy 细菌纳米颗粒作为一种有效和安全的替代卡介苗用于膀胱癌免疫治疗。
IF 5 3区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2025-10-30 DOI: 10.1038/s41417-025-00984-8
Marine Colson, Arthur Aubry, Nicolas Gaide, Philippe Lluel, Eric Oswald, Laure David
Bladder cancer is the second most common malignancy of the urogenital system, with non-muscle-invasive forms being the most prevalent. While therapy with intravesical instillation of Bacillus Calmette-Guérin (BCG) is widely used, it shows variable efficacy and notable side effects, which prompts the need for new treatments. This study introduces a nanoscale biotherapy against bladder cancer using bacterial extracellular vesicles (bEVs) from Escherichia coli. In an orthotopic syngeneic rat model of bladder cancer with luciferase-tagged tumour cells, intravesical bEVs treatment significantly inhibited tumour growth compared to the gold standard therapy: BCG. Indeed, compared to BCG, bEVs treatment led to superior tumour regression, with 60% of treated animals showing complete tumour clearance versus 20% in the BCG-treated group. Moreover, we observed strong local immune activation, namely T cell infiltration and mucosal lymphoid aggregates, which negatively correlated with tumour size, suggesting that bEVs efficacy is due, at least in part, to their ability to induce a strong antitumor immune response. Notably, bEVs caused no mortality or systemic toxicity. Altogether, the present study constitutes a striking illustration of translational medicine, where the properties of a bacterial compound can be harnessed for patient benefit.
膀胱癌是泌尿生殖系统第二常见的恶性肿瘤,非肌肉侵袭性形式是最普遍的。虽然膀胱内注射卡介苗(Bacillus calmetet - gusamrin, BCG)治疗被广泛使用,但其疗效不一且副作用明显,这促使人们需要新的治疗方法。本研究介绍了一种利用大肠杆菌细菌细胞外囊泡(bEVs)治疗膀胱癌的纳米级生物疗法。在具有荧光素酶标记肿瘤细胞的原位同基因膀胱癌大鼠模型中,与金标准疗法卡介苗相比,膀胱内bEVs治疗显著抑制肿瘤生长。事实上,与BCG相比,bEVs治疗导致了更好的肿瘤消退,60%的治疗动物显示完全的肿瘤清除,而BCG治疗组为20%。此外,我们观察到强烈的局部免疫激活,即T细胞浸润和粘膜淋巴细胞聚集,这与肿瘤大小呈负相关,这表明bev的功效至少部分归因于它们诱导强烈的抗肿瘤免疫反应的能力。值得注意的是,纯电动汽车没有造成死亡或全身毒性。总的来说,目前的研究构成了转化医学的一个引人注目的例证,其中细菌化合物的特性可以被利用来造福患者。
{"title":"Bacterial nanoparticles as a potent and safe alternative to BCG for bladder cancer immunotherapy","authors":"Marine Colson,&nbsp;Arthur Aubry,&nbsp;Nicolas Gaide,&nbsp;Philippe Lluel,&nbsp;Eric Oswald,&nbsp;Laure David","doi":"10.1038/s41417-025-00984-8","DOIUrl":"10.1038/s41417-025-00984-8","url":null,"abstract":"Bladder cancer is the second most common malignancy of the urogenital system, with non-muscle-invasive forms being the most prevalent. While therapy with intravesical instillation of Bacillus Calmette-Guérin (BCG) is widely used, it shows variable efficacy and notable side effects, which prompts the need for new treatments. This study introduces a nanoscale biotherapy against bladder cancer using bacterial extracellular vesicles (bEVs) from Escherichia coli. In an orthotopic syngeneic rat model of bladder cancer with luciferase-tagged tumour cells, intravesical bEVs treatment significantly inhibited tumour growth compared to the gold standard therapy: BCG. Indeed, compared to BCG, bEVs treatment led to superior tumour regression, with 60% of treated animals showing complete tumour clearance versus 20% in the BCG-treated group. Moreover, we observed strong local immune activation, namely T cell infiltration and mucosal lymphoid aggregates, which negatively correlated with tumour size, suggesting that bEVs efficacy is due, at least in part, to their ability to induce a strong antitumor immune response. Notably, bEVs caused no mortality or systemic toxicity. Altogether, the present study constitutes a striking illustration of translational medicine, where the properties of a bacterial compound can be harnessed for patient benefit.","PeriodicalId":9577,"journal":{"name":"Cancer gene therapy","volume":"33 1","pages":"76-84"},"PeriodicalIF":5.0,"publicationDate":"2025-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145408083","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Cancer gene therapy
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1