首页 > 最新文献

Oncogene最新文献

英文 中文
Retraction Note: Claudin-1 induces epithelial–mesenchymal transition through activation of the c-Abl-ERK signaling pathway in human liver cells 注:cludin -1通过激活人肝细胞c-Abl-ERK信号通路诱导上皮-间质转化。
IF 7.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-09 DOI: 10.1038/s41388-025-03673-w
Y. Suh, C-H Yoon, R-K Kim, E-J Lim, YS Oh, S-G Hwang, S. An, G. Yoon, M. C. Gye, J-M Yi, M-J Kim, S-J Lee
{"title":"Retraction Note: Claudin-1 induces epithelial–mesenchymal transition through activation of the c-Abl-ERK signaling pathway in human liver cells","authors":"Y. Suh, C-H Yoon, R-K Kim, E-J Lim, YS Oh, S-G Hwang, S. An, G. Yoon, M. C. Gye, J-M Yi, M-J Kim, S-J Lee","doi":"10.1038/s41388-025-03673-w","DOIUrl":"10.1038/s41388-025-03673-w","url":null,"abstract":"","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":"45 4","pages":"591-591"},"PeriodicalIF":7.3,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.comhttps://www.nature.com/articles/s41388-025-03673-w.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145945607","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}
引用次数: 0
BAP1-loss in mesothelioma: molecular mechanisms and clinical opportunities. 间皮瘤中bap1的丢失:分子机制和临床机会。
IF 7.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-07 DOI: 10.1038/s41388-025-03672-x
Jasper H L T van Genugten, Dean A Fennell, Paul Baas

Mesothelioma is an aggressive cancer that is often characterized by loss of the BRCA1-associated protein 1 (BAP1) tumor suppressor gene. This alteration typically occurs as an early clonal event in mesothelioma development, making it a promising candidate for both diagnostic and therapeutic applications. Functionally, BAP1 regulates gene expression through interactions with Polycomb-group complexes, and it plays roles in various other cellular processes including DNA repair, replication stress, and cell metabolism. While preclinical research has identified multiple potential vulnerabilities in BAP1-deficient tumors-including sensitivity to EZH2-, HDAC-, PARP-, and FGFR-inhibitors-translating these findings to the clinic remains a challenge. In this review, we provide a comprehensive overview of BAP1's molecular functions in mesothelioma, with a focus on their translation into clinical therapeutics for this hard-to-treat malignancy.

间皮瘤是一种侵袭性癌症,通常以brca1相关蛋白1 (BAP1)肿瘤抑制基因的缺失为特征。这种改变通常发生在间皮瘤发展的早期克隆事件中,使其成为诊断和治疗应用的有希望的候选者。在功能上,BAP1通过与Polycomb-group复合物的相互作用调节基因表达,并在DNA修复、复制应激和细胞代谢等多种细胞过程中发挥作用。虽然临床前研究已经确定了bap1缺陷肿瘤的多种潜在脆弱性,包括对EZH2-、HDAC-、PARP-和fgfr -抑制剂的敏感性,但将这些发现转化为临床仍然是一个挑战。在这篇综述中,我们提供了BAP1在间皮瘤中的分子功能的全面概述,重点是它们在这种难以治疗的恶性肿瘤的临床治疗中的转化。
{"title":"BAP1-loss in mesothelioma: molecular mechanisms and clinical opportunities.","authors":"Jasper H L T van Genugten, Dean A Fennell, Paul Baas","doi":"10.1038/s41388-025-03672-x","DOIUrl":"https://doi.org/10.1038/s41388-025-03672-x","url":null,"abstract":"<p><p>Mesothelioma is an aggressive cancer that is often characterized by loss of the BRCA1-associated protein 1 (BAP1) tumor suppressor gene. This alteration typically occurs as an early clonal event in mesothelioma development, making it a promising candidate for both diagnostic and therapeutic applications. Functionally, BAP1 regulates gene expression through interactions with Polycomb-group complexes, and it plays roles in various other cellular processes including DNA repair, replication stress, and cell metabolism. While preclinical research has identified multiple potential vulnerabilities in BAP1-deficient tumors-including sensitivity to EZH2-, HDAC-, PARP-, and FGFR-inhibitors-translating these findings to the clinic remains a challenge. In this review, we provide a comprehensive overview of BAP1's molecular functions in mesothelioma, with a focus on their translation into clinical therapeutics for this hard-to-treat malignancy.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145918228","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}
引用次数: 0
EZH2 PROTACs outperform catalytic inhibitors in prostate cancer by targeting a methylation-independent function of PRC2. EZH2 PROTACs通过靶向PRC2的甲基化非依赖性功能,在前列腺癌治疗中优于催化抑制剂。
IF 7.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-07 DOI: 10.1038/s41388-025-03662-z
Wanqing Xie, Qi Chu, Lourdes Brea, Guihua Zeng, Yuan Wang, Xiaodong Lu, Mohan Zheng, Corinne R Ley, Zhiquan Lei, Hongshun Shi, Joshua L Zhu, Lihu Gong, M Cynthia Martin, Xianglin Shi, Galina Gritsina, Arabela A Grigorescu, Hana Chandonnet, Xin Liu, Jonathan C Zhao, Gary E Schiltz, Jindan Yu

Enhancer of Zeste Homolog 2 (EZH2) is the enzymatic subunit of the Polycomb Repressive Complex 2 (PRC2). It catalyzes H3K27 methylation for epigenetic silencing of tumor suppressors and critically drives prostate cancer (PCa) progression. However, inhibitors of EZH2 catalytic function (EZH2i), such as EPZ-6438, showed limited efficacy in PCa. Here, we designed and developed a series of VHL-based proteolysis-targeting chimera (PROTAC) degraders of EZH2 using EPZ-6438 as a ligand and identified PROTAC-6272 as a lead compound. PROTAC-6272 effectively degraded EZH2 and other PRC2 subunits across diverse PCa cell lines. However, PROTAC-6272 and other similar EZH2i-based PROTACs were consistently unable to decrease androgen receptor (AR), a gene that is directly activated by solo EZH2. Mechanistically, EZH2 PROTACs failed to degrade EZH2 coactivators, such as p300, due to their inability to engage EZH2 outside of the PRC2 complex. Nevertheless, PROTAC-6272 exhibited anti-proliferative activities superior to EPZ-6438 in some PCa models, wherein it induced p21 expression and cellular senescence by disrupting a methylation-independent PRC2 function. In summary, while EZH2i-based PROTACs failed to target the PRC2-independent functions of EZH2, they confer added benefits over EPZ-6438 by abolishing a polycomb-dependent but methylation-independent function of EZH2, offering therapeutic advantages in some PCa.

Zeste同源物2增强子(EZH2)是多梳抑制复合物2 (PRC2)的酶亚基。它催化H3K27甲基化,使肿瘤抑制因子表观遗传沉默,并关键地驱动前列腺癌(PCa)的进展。然而,EZH2催化功能抑制剂(EZH2i),如EPZ-6438,对PCa的疗效有限。本研究以EPZ-6438为配体,设计并开发了一系列基于vhl的EZH2蛋白水解靶向嵌合体(PROTAC)降解物,并鉴定了PROTAC-6272为先导化合物。PROTAC-6272在多种PCa细胞系中有效降解EZH2和其他PRC2亚基。然而,PROTAC-6272和其他类似的基于EZH2的PROTACs始终不能降低雄激素受体(AR),这是一种由单独EZH2直接激活的基因。从机制上讲,EZH2 PROTACs无法降解EZH2共激活因子,如p300,因为它们无法与PRC2复合物外的EZH2结合。然而,在一些PCa模型中,PROTAC-6272表现出优于EPZ-6438的抗增殖活性,其中PROTAC-6272通过破坏甲基化无关的PRC2功能诱导p21表达和细胞衰老。综上所述,尽管基于ezh2i的PROTACs无法靶向EZH2的prc2独立功能,但它们通过消除EZH2依赖于polycomb但不依赖于甲基化的功能,赋予EPZ-6438额外的益处,从而在某些PCa中提供治疗优势。
{"title":"EZH2 PROTACs outperform catalytic inhibitors in prostate cancer by targeting a methylation-independent function of PRC2.","authors":"Wanqing Xie, Qi Chu, Lourdes Brea, Guihua Zeng, Yuan Wang, Xiaodong Lu, Mohan Zheng, Corinne R Ley, Zhiquan Lei, Hongshun Shi, Joshua L Zhu, Lihu Gong, M Cynthia Martin, Xianglin Shi, Galina Gritsina, Arabela A Grigorescu, Hana Chandonnet, Xin Liu, Jonathan C Zhao, Gary E Schiltz, Jindan Yu","doi":"10.1038/s41388-025-03662-z","DOIUrl":"https://doi.org/10.1038/s41388-025-03662-z","url":null,"abstract":"<p><p>Enhancer of Zeste Homolog 2 (EZH2) is the enzymatic subunit of the Polycomb Repressive Complex 2 (PRC2). It catalyzes H3K27 methylation for epigenetic silencing of tumor suppressors and critically drives prostate cancer (PCa) progression. However, inhibitors of EZH2 catalytic function (EZH2i), such as EPZ-6438, showed limited efficacy in PCa. Here, we designed and developed a series of VHL-based proteolysis-targeting chimera (PROTAC) degraders of EZH2 using EPZ-6438 as a ligand and identified PROTAC-6272 as a lead compound. PROTAC-6272 effectively degraded EZH2 and other PRC2 subunits across diverse PCa cell lines. However, PROTAC-6272 and other similar EZH2i-based PROTACs were consistently unable to decrease androgen receptor (AR), a gene that is directly activated by solo EZH2. Mechanistically, EZH2 PROTACs failed to degrade EZH2 coactivators, such as p300, due to their inability to engage EZH2 outside of the PRC2 complex. Nevertheless, PROTAC-6272 exhibited anti-proliferative activities superior to EPZ-6438 in some PCa models, wherein it induced p21 expression and cellular senescence by disrupting a methylation-independent PRC2 function. In summary, while EZH2i-based PROTACs failed to target the PRC2-independent functions of EZH2, they confer added benefits over EPZ-6438 by abolishing a polycomb-dependent but methylation-independent function of EZH2, offering therapeutic advantages in some PCa.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145918172","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}
引用次数: 0
An H4K12la/CEBPB-AKR1C2 signaling axis modulates the mTOR pathway to regulate cisplatin resistance in lung cancer. H4K12la/CEBPB-AKR1C2信号轴调控mTOR通路调控肺癌顺铂耐药
IF 7.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-07 DOI: 10.1038/s41388-025-03669-6
Wenjing Wang, Qiang He, Tianfei Fan, Yang Xiong, Yimin Xiong, Qinhui Liu, Na Yang, Yining Xu, Yanping Li, Jinhan He

Histone lactylation, a recently discovered epigenetic modification, has been shown to play a critical role in regulating gene expression and cellular functions. However, its involvement in cisplatin (CDDP) resistance in non-small cell lung cancer (NSCLC) remains poorly understood. In this study, we demonstrated that histone lactylation is closely associated with CDDP resistance and correlates with poor prognosis of NSCLC. Mechanistically, H4K12la (histone e 4 lysine 12 lactylation) levels and CEBPB (CCAAT/enhancer-binding protein beta) had a cooperative effect on the regulation of AKR1C2 (Aldo-Keto reductase 1C2). Furthermore, AKR1C2 knockdown activates the mTOR oncogenic signaling pathway. Importantly, genetic manipulation of AKR1C2 or the combination of CDDP with an mTOR inhibitor effectively reverse CDDP resistance in NSCLC/CDDP cells. These findings highlighted the potential of AKR1C2 as a predictive biomarker for patient response to CDDP therapy. Additionally, our study established a novel link between histone lactylation and CDDP resistance, providing new insights into the epigenetic regulation in NSCLC.

组蛋白乳酸化是最近发现的一种表观遗传修饰,在调节基因表达和细胞功能方面起着关键作用。然而,它在非小细胞肺癌(NSCLC)顺铂(CDDP)耐药中的作用仍然知之甚少。在本研究中,我们证实组蛋白乳酸化与CDDP耐药密切相关,并与NSCLC的不良预后相关。机制上,H4K12la(组蛋白4赖氨酸12乳酸化)水平和CEBPB (CCAAT/增强子结合蛋白β)协同作用于AKR1C2 (Aldo-Keto还原酶1C2)的调控。此外,AKR1C2敲低激活mTOR致癌信号通路。重要的是,AKR1C2的基因操作或CDDP与mTOR抑制剂的联合有效地逆转了NSCLC/CDDP细胞中的CDDP耐药性。这些发现强调了AKR1C2作为预测患者对CDDP治疗反应的生物标志物的潜力。此外,我们的研究建立了组蛋白乳酸化与CDDP耐药之间的新联系,为非小细胞肺癌的表观遗传调控提供了新的见解。
{"title":"An H4K12la/CEBPB-AKR1C2 signaling axis modulates the mTOR pathway to regulate cisplatin resistance in lung cancer.","authors":"Wenjing Wang, Qiang He, Tianfei Fan, Yang Xiong, Yimin Xiong, Qinhui Liu, Na Yang, Yining Xu, Yanping Li, Jinhan He","doi":"10.1038/s41388-025-03669-6","DOIUrl":"https://doi.org/10.1038/s41388-025-03669-6","url":null,"abstract":"<p><p>Histone lactylation, a recently discovered epigenetic modification, has been shown to play a critical role in regulating gene expression and cellular functions. However, its involvement in cisplatin (CDDP) resistance in non-small cell lung cancer (NSCLC) remains poorly understood. In this study, we demonstrated that histone lactylation is closely associated with CDDP resistance and correlates with poor prognosis of NSCLC. Mechanistically, H4K12la (histone e 4 lysine 12 lactylation) levels and CEBPB (CCAAT/enhancer-binding protein beta) had a cooperative effect on the regulation of AKR1C2 (Aldo-Keto reductase 1C2). Furthermore, AKR1C2 knockdown activates the mTOR oncogenic signaling pathway. Importantly, genetic manipulation of AKR1C2 or the combination of CDDP with an mTOR inhibitor effectively reverse CDDP resistance in NSCLC/CDDP cells. These findings highlighted the potential of AKR1C2 as a predictive biomarker for patient response to CDDP therapy. Additionally, our study established a novel link between histone lactylation and CDDP resistance, providing new insights into the epigenetic regulation in NSCLC.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145918241","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}
引用次数: 0
Targeting STING elicits GSDMD-dependent pyroptosis and boosts anti-tumor immunity in renal cell carcinoma. 靶向STING诱导gsdmd依赖性焦亡并增强肾细胞癌的抗肿瘤免疫。
IF 7.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-04 DOI: 10.1038/s41388-025-03671-y
Shengpan Wu, Baojun Wang, Hongzhao Li, Hanfeng Wang, Songliang Du, Xing Huang, Yang Fan, Yu Gao, Liangyou Gu, Qingbo Huang, Jianjun Chen, Xu Zhang, Yan Huang, Xin Ma

While Stimulator-of-interferon genes (STING) is an innate immune adapter crucial for sensing cytosolic DNA and modulating immune microenvironment, its tumor-promoting role in tumor survival and immune evasion remains largely unknown. Here we reported that renal cancer cells are exceptionally dependent on STING for survival and evading immunosurveillance via suppressing ER stress-mediated pyroptosis. We found that STING is significantly amplified and upregulated in clear cell renal cell carcinoma (ccRCC), and its elevated expression is associated with worse clinical outcomes. Mechanically, STING depletion in RCC cells specifically triggers activation of the PERK/eIF2α/ATF4/CHOP pathway and activates cleavage of Caspase-8, thereby inducing GSDMD-mediated pyroptosis, which is independent of the innate immune pathway of STING. Moreover, animal study results revealed that STING depletion promoted infiltration of CD4+ and CD8+ T cells, consequently boosting robust antitumor immunity via pyroptosis-induced inflammation. From the perspective of targeted therapy, we found that Compound SP23, a PROTAC STING degrader, demonstrated comparable efficacy to STING depletion both in vitro and in vivo for treatment of ccRCC. These findings collectively unveiled an unforeseen function of STING in regulating GSDMD-dependent pyroptosis, thus regulating immune response in RCC. Consequently, pharmacological degradation of STING by SP23 may become an attractive strategy for treatment of advanced RCC.

虽然干扰素刺激因子基因(STING)是一种先天免疫适配器,对感知胞质DNA和调节免疫微环境至关重要,但其在肿瘤存活和免疫逃避中的促肿瘤作用仍未得到充分的了解。在这里,我们报道了肾癌细胞异常依赖STING生存,并通过抑制内质网应激介导的焦亡来逃避免疫监视。我们发现,STING在透明细胞肾细胞癌(ccRCC)中显著扩增和上调,其表达升高与较差的临床结果相关。机械上,RCC细胞中STING的缺失特异性触发PERK/eIF2α/ATF4/CHOP通路的激活,激活Caspase-8的裂解,从而诱导gsdmd介导的焦亡,而这与STING的先天免疫通路无关。此外,动物研究结果显示,STING耗竭促进CD4+和CD8+ T细胞的浸润,从而通过焦热诱导的炎症增强强大的抗肿瘤免疫。从靶向治疗的角度来看,我们发现复方SP23,一种PROTAC STING降解剂,在体外和体内治疗ccRCC的疗效与STING耗尽相当。这些发现共同揭示了STING在调节gsdmd依赖性焦亡中的一个意想不到的功能,从而调节RCC的免疫反应。因此,SP23对STING的药理降解可能成为治疗晚期RCC的一种有吸引力的策略。
{"title":"Targeting STING elicits GSDMD-dependent pyroptosis and boosts anti-tumor immunity in renal cell carcinoma.","authors":"Shengpan Wu, Baojun Wang, Hongzhao Li, Hanfeng Wang, Songliang Du, Xing Huang, Yang Fan, Yu Gao, Liangyou Gu, Qingbo Huang, Jianjun Chen, Xu Zhang, Yan Huang, Xin Ma","doi":"10.1038/s41388-025-03671-y","DOIUrl":"https://doi.org/10.1038/s41388-025-03671-y","url":null,"abstract":"<p><p>While Stimulator-of-interferon genes (STING) is an innate immune adapter crucial for sensing cytosolic DNA and modulating immune microenvironment, its tumor-promoting role in tumor survival and immune evasion remains largely unknown. Here we reported that renal cancer cells are exceptionally dependent on STING for survival and evading immunosurveillance via suppressing ER stress-mediated pyroptosis. We found that STING is significantly amplified and upregulated in clear cell renal cell carcinoma (ccRCC), and its elevated expression is associated with worse clinical outcomes. Mechanically, STING depletion in RCC cells specifically triggers activation of the PERK/eIF2α/ATF4/CHOP pathway and activates cleavage of Caspase-8, thereby inducing GSDMD-mediated pyroptosis, which is independent of the innate immune pathway of STING. Moreover, animal study results revealed that STING depletion promoted infiltration of CD4<sup>+</sup> and CD8<sup>+</sup> T cells, consequently boosting robust antitumor immunity via pyroptosis-induced inflammation. From the perspective of targeted therapy, we found that Compound SP23, a PROTAC STING degrader, demonstrated comparable efficacy to STING depletion both in vitro and in vivo for treatment of ccRCC. These findings collectively unveiled an unforeseen function of STING in regulating GSDMD-dependent pyroptosis, thus regulating immune response in RCC. Consequently, pharmacological degradation of STING by SP23 may become an attractive strategy for treatment of advanced RCC.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2026-01-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145896709","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}
引用次数: 0
The role of the SLC25A15 transporter in the formation of liver metastasis in ESR1-mutated breast cancer. SLC25A15转运体在esr1突变乳腺癌肝转移形成中的作用
IF 7.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2026-01-03 DOI: 10.1038/s41388-025-03640-5
Marwa Taya, Daniel Fishman, Fahim Kanani, Lotem Zinger, Keren Merenbakh-Lamin, Shaked Elfasi Sosner, Anil Khushalrao Shendge, Anat Klein Goldberg, Uri Wolf, Galit Winkler, Eric Shifrut, Shmuel Cohen, Ido Wolf, Israel Sekler, Tami Rubinek

Activating mutations in the ligand-binding domain of the estrogen receptor (ER)-encoding (ESR1) gene are present in up to 40% of metastatic breast cancer (BC) patients and are strongly associated with a high risk of liver metastasis (LM) formation. Using the MCF-7 BC model, we investigated whether the increased hepatic tropism of ESR1-mutated BC cells is driven by their metabolic adaptation to the liver microenvironment. Indeed, metabolomic analysis revealed elevated metabolites related to the urea cycle (UC) in LM-forming ESR1-mutated cells compared to wild-type (WT) ER-expressing cells, which failed to generate LM. The subsequent proteomic, western blotting, and qPCR analyses demonstrated a dramatic upregulation of the UC constituent, the mitochondrial ornithine/citrulline transporter SLC25A15, in liver-predilected ESR1-mutated cells relative to their WT counterpart cells. Unlike WT cells, ESR1-mutated cells readily formed spheroids and exhibited enhanced migration in liver mimicking hepatocyte-conditioned media. In addition, we employed a novel ex vivo approach where ESR1 mutated cells were seeded onto colonized fresh liver tissue-which was abolished by SLC25A15 knockout. Moreover, SLC25A15 knockout robustly reduced the ability of ESR1-mutated cells to establish LM in vivo. These findings highlight SLC25A15-mediated dysregulation of the UC as a critical driver of BC hepatic metastasis and identify SLC25A15 as a potential therapeutic target for disrupting metastatic spread of BC to the liver.

雌激素受体(ER)编码(ESR1)基因配体结合域的激活突变存在于高达40%的转移性乳腺癌(BC)患者中,并且与肝转移(LM)形成的高风险密切相关。使用MCF-7 BC模型,我们研究了esr1突变的BC细胞的肝向性增加是否由它们对肝脏微环境的代谢适应驱动。事实上,代谢组学分析显示,与野生型(WT) er表达细胞相比,形成LM的esr1突变细胞中与尿素循环(UC)相关的代谢物升高,而野生型(WT) er表达细胞无法产生LM。随后的蛋白质组学、western blotting和qPCR分析表明,与WT对应的细胞相比,肝脏选择性esr1突变细胞中的UC成分、线粒体鸟氨酸/瓜氨酸转运体SLC25A15显著上调。与WT细胞不同,esr1突变的细胞在模拟肝细胞条件的培养基中容易形成球状体并表现出增强的迁移。此外,我们采用了一种新颖的离体方法,将ESR1突变细胞播种到定植的新鲜肝组织中,通过敲除SLC25A15来消除。此外,SLC25A15敲除显著降低了esr1突变细胞在体内建立LM的能力。这些发现强调了SLC25A15介导的UC失调是BC肝转移的关键驱动因素,并确定SLC25A15是破坏BC转移到肝脏的潜在治疗靶点。
{"title":"The role of the SLC25A15 transporter in the formation of liver metastasis in ESR1-mutated breast cancer.","authors":"Marwa Taya, Daniel Fishman, Fahim Kanani, Lotem Zinger, Keren Merenbakh-Lamin, Shaked Elfasi Sosner, Anil Khushalrao Shendge, Anat Klein Goldberg, Uri Wolf, Galit Winkler, Eric Shifrut, Shmuel Cohen, Ido Wolf, Israel Sekler, Tami Rubinek","doi":"10.1038/s41388-025-03640-5","DOIUrl":"https://doi.org/10.1038/s41388-025-03640-5","url":null,"abstract":"<p><p>Activating mutations in the ligand-binding domain of the estrogen receptor (ER)-encoding (ESR1) gene are present in up to 40% of metastatic breast cancer (BC) patients and are strongly associated with a high risk of liver metastasis (LM) formation. Using the MCF-7 BC model, we investigated whether the increased hepatic tropism of ESR1-mutated BC cells is driven by their metabolic adaptation to the liver microenvironment. Indeed, metabolomic analysis revealed elevated metabolites related to the urea cycle (UC) in LM-forming ESR1-mutated cells compared to wild-type (WT) ER-expressing cells, which failed to generate LM. The subsequent proteomic, western blotting, and qPCR analyses demonstrated a dramatic upregulation of the UC constituent, the mitochondrial ornithine/citrulline transporter SLC25A15, in liver-predilected ESR1-mutated cells relative to their WT counterpart cells. Unlike WT cells, ESR1-mutated cells readily formed spheroids and exhibited enhanced migration in liver mimicking hepatocyte-conditioned media. In addition, we employed a novel ex vivo approach where ESR1 mutated cells were seeded onto colonized fresh liver tissue-which was abolished by SLC25A15 knockout. Moreover, SLC25A15 knockout robustly reduced the ability of ESR1-mutated cells to establish LM in vivo. These findings highlight SLC25A15-mediated dysregulation of the UC as a critical driver of BC hepatic metastasis and identify SLC25A15 as a potential therapeutic target for disrupting metastatic spread of BC to the liver.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2026-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145896755","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}
引用次数: 0
PDZRN4 suppresses lung adenocarcinoma progression via inhibiting ubiquitin-mediated HIF-1A degradation PDZRN4通过抑制泛素介导的HIF-1A降解来抑制肺腺癌的进展。
IF 7.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-29 DOI: 10.1038/s41388-025-03670-z
Honghao Li, Junfeng Wu, Hua Dai, Jiaming Li, Xiaoyan Bai, Yueli Sun, Jiaxin Lin, Yangsi Li, Peng Li, Lingchao Shi, Zhen Wang, Haiyan Tu, Ee Ke
Indolent lung cancer is characterized by slow progression, and the patient may die of other causes before it becomes symptomatic. The increasing usage of lung cancer screening technologies has increased the detection of early-stage cancers, improved survival, and reduced mortality, but it also brings overdiagnosis or overtreatment for indolent lung cancer patients. In this study, we performed high-throughput whole-exome sequencing in lung adenocarcinoma (LUAD) patients with a tumor doubling time of more than 600 days (the indolent group) and another group with a tumor doubling time of less than 30 days (the aggressive group). We identified a novel germline variation in PDZRN4, rs74955204 (p.G121E, c.362 G > A) in the indolent group. The rs74955204 variant resulted in upregulated PDZRN4 expression and was positively correlated with the survival of LUAD patients. We confirmed that overexpression of PDZRN4 inhibits the ubiquitylation of HIF-1A mediated by TRIM28, thus activating HIF-1A-IGFBP3 signaling to promote apoptosis, and inducing NDRG1 to inhibit EGFR-AKT signaling, restricting lung cancer cell growth in vitro and in vivo. With the EGFRL858R-driven lung cancer mouse model as an example, we found that PDZRN4 knockout promoted, whereas PDZRN4 overexpression suppressed LUAD growth in vivo. In addition, recombinant PDZRN4 or screened small molecules retinoic acid showed potent inhibitory effects on EGFR-driven LUAD growth and amelioration of osimertinib resistance. Overall, our findings suggest that targeted PDZRN4 is a potential therapeutic strategy for treating LUAD. Examination of PDZRN4 rs74955204 variant may be an effective approach for the early detection of indolent lung cancer and to avoid overdiagnosis.
惰性肺癌的特点是进展缓慢,患者可能在出现症状之前死于其他原因。肺癌筛查技术的日益普及,增加了早期癌症的发现,提高了生存率,降低了死亡率,但也给惰性肺癌患者带来了过度诊断或过度治疗。在本研究中,我们对肿瘤加倍时间超过600天的肺腺癌(LUAD)患者(惰性组)和肿瘤加倍时间小于30天的肺腺癌(LUAD)患者(侵袭组)进行了高通量全外显子组测序。我们在惰性组中发现了PDZRN4, rs74955204 (p.G121E, c.362 G > a)的一种新的种系变异。rs74955204变异导致PDZRN4表达上调,与LUAD患者的生存呈正相关。我们在体外和体内证实,PDZRN4过表达可抑制TRIM28介导的HIF-1A泛素化,从而激活HIF-1A- igfbp3信号通路促进细胞凋亡,诱导NDRG1抑制EGFR-AKT信号通路,限制肺癌细胞生长。以egfrl858r驱动的肺癌小鼠模型为例,我们发现PDZRN4敲除促进体内LUAD的生长,而PDZRN4过表达抑制体内LUAD的生长。此外,重组PDZRN4或筛选的小分子维甲酸对egfr驱动的LUAD生长和改善奥西替尼耐药性有明显的抑制作用。总之,我们的研究结果表明,靶向PDZRN4是治疗LUAD的潜在治疗策略。检测PDZRN4 rs74955204变异可能是早期发现惰性肺癌,避免过度诊断的有效途径。摘要PDZRN4的种系变异rs74955204 (p.G121E, c.g 362 G > A)显著上调PDZRN4的表达,导致LUAD的惰性生长。过表达的PDZRN4可抑制TRIM28对HIF-1A的泛素化,激活HIF-1A- igfbp3信号通路促进细胞凋亡,诱导NDRG1抑制EGFR-AKT信号通路,从而限制肺癌细胞生长。
{"title":"PDZRN4 suppresses lung adenocarcinoma progression via inhibiting ubiquitin-mediated HIF-1A degradation","authors":"Honghao Li,&nbsp;Junfeng Wu,&nbsp;Hua Dai,&nbsp;Jiaming Li,&nbsp;Xiaoyan Bai,&nbsp;Yueli Sun,&nbsp;Jiaxin Lin,&nbsp;Yangsi Li,&nbsp;Peng Li,&nbsp;Lingchao Shi,&nbsp;Zhen Wang,&nbsp;Haiyan Tu,&nbsp;Ee Ke","doi":"10.1038/s41388-025-03670-z","DOIUrl":"10.1038/s41388-025-03670-z","url":null,"abstract":"Indolent lung cancer is characterized by slow progression, and the patient may die of other causes before it becomes symptomatic. The increasing usage of lung cancer screening technologies has increased the detection of early-stage cancers, improved survival, and reduced mortality, but it also brings overdiagnosis or overtreatment for indolent lung cancer patients. In this study, we performed high-throughput whole-exome sequencing in lung adenocarcinoma (LUAD) patients with a tumor doubling time of more than 600 days (the indolent group) and another group with a tumor doubling time of less than 30 days (the aggressive group). We identified a novel germline variation in PDZRN4, rs74955204 (p.G121E, c.362 G &gt; A) in the indolent group. The rs74955204 variant resulted in upregulated PDZRN4 expression and was positively correlated with the survival of LUAD patients. We confirmed that overexpression of PDZRN4 inhibits the ubiquitylation of HIF-1A mediated by TRIM28, thus activating HIF-1A-IGFBP3 signaling to promote apoptosis, and inducing NDRG1 to inhibit EGFR-AKT signaling, restricting lung cancer cell growth in vitro and in vivo. With the EGFRL858R-driven lung cancer mouse model as an example, we found that PDZRN4 knockout promoted, whereas PDZRN4 overexpression suppressed LUAD growth in vivo. In addition, recombinant PDZRN4 or screened small molecules retinoic acid showed potent inhibitory effects on EGFR-driven LUAD growth and amelioration of osimertinib resistance. Overall, our findings suggest that targeted PDZRN4 is a potential therapeutic strategy for treating LUAD. Examination of PDZRN4 rs74955204 variant may be an effective approach for the early detection of indolent lung cancer and to avoid overdiagnosis.","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":"45 4","pages":"577-590"},"PeriodicalIF":7.3,"publicationDate":"2025-12-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145857129","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}
引用次数: 0
NAT10 triggers colorectal cancer progression via promoting PPAN-regulated DNA damage repair NAT10通过促进ppan调节的DNA损伤修复来触发结直肠癌的进展。
IF 7.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-27 DOI: 10.1038/s41388-025-03661-0
Haoran Wang, Lichen Ge, Jianing Li, Ke Zhong, Shanzhi Li, Ningjing Ma, Lijun Tao, Jiawang Zhou, Zhaotong Wang, Xing Chang, Yunqing Lu, Yalan Rui, Guoyou Xie, Weifeng Yang, Zuanzong Xu, Abdulaziz Ahmed A. Saad, Xiansong Wang, Zhuojia Chen, Wanglin Li, Cheng Yi, Hongsheng Wang, Kun Zhang
RNA modification recognition proteins are crucial in cancer development and progression. Among all RNA modification-related proteins (RMRPs), Weighted Gene Co-expression Network Analysis (WGCNA), combined with comprehensive bioinformatic analysis, suggests that NAT10—the sole known writer of N4-acetylcytidine (ac4C)—is a critical regulatory protein in colorectal cancer (CRC) progression. NAT10 facilitates the malignancy phenotypes and DNA damage repair in CRC cells via its ac4C transferase activity and regulation of PPAN. Specifically, NAT10 enhances the translation efficiency of PPAN via acetylation at the C744 and C747 sites. In addition, NAT10 promotes the translation of ac4C-modified MYC mRNA. MYC protein then enhances PPAN transcription through binding to the PPAN promoter. The newly identified ac4C reader protein MYBBP1A mediates NAT10-induced translation of both PPAN and MYC. We further found that VDR binds to the NAT10 promoter to activate its transcription, resulting in the high expression of NAT10 in CRC. Xenograft studies and clinical data confirmed the role of the NAT10-PPAN axis in promoting CRC development and DNA damage repair. Collectively, our study reveals the role and underlying mechanism of mRNA ac4C modification in CRC progression, providing critical potential targets for CRC drug development.
RNA修饰识别蛋白在癌症的发生和发展中是至关重要的。在所有RNA修饰相关蛋白(RMRPs)中,加权基因共表达网络分析(WGCNA)结合综合生物信息学分析表明,n4 -乙酰胞苷(ac4C)的唯一已知作者nat10是结直肠癌(CRC)进展的关键调控蛋白。NAT10通过其ac4C转移酶活性和对PPAN的调节促进CRC细胞的恶性表型和DNA损伤修复。具体来说,NAT10通过在C744和C747位点乙酰化来提高PPAN的翻译效率。此外,NAT10促进ac4c修饰的MYC mRNA的翻译。然后MYC蛋白通过结合PPAN启动子来增强PPAN的转录。新发现的ac4C解读蛋白MYBBP1A介导nat10诱导的PPAN和MYC的翻译。我们进一步发现VDR与NAT10启动子结合激活其转录,导致NAT10在CRC中高表达。异种移植研究和临床数据证实了NAT10-PPAN轴在促进结直肠癌发展和DNA损伤修复中的作用。总之,我们的研究揭示了mRNA ac4C修饰在结直肠癌进展中的作用和潜在机制,为结直肠癌药物开发提供了关键的潜在靶点。
{"title":"NAT10 triggers colorectal cancer progression via promoting PPAN-regulated DNA damage repair","authors":"Haoran Wang,&nbsp;Lichen Ge,&nbsp;Jianing Li,&nbsp;Ke Zhong,&nbsp;Shanzhi Li,&nbsp;Ningjing Ma,&nbsp;Lijun Tao,&nbsp;Jiawang Zhou,&nbsp;Zhaotong Wang,&nbsp;Xing Chang,&nbsp;Yunqing Lu,&nbsp;Yalan Rui,&nbsp;Guoyou Xie,&nbsp;Weifeng Yang,&nbsp;Zuanzong Xu,&nbsp;Abdulaziz Ahmed A. Saad,&nbsp;Xiansong Wang,&nbsp;Zhuojia Chen,&nbsp;Wanglin Li,&nbsp;Cheng Yi,&nbsp;Hongsheng Wang,&nbsp;Kun Zhang","doi":"10.1038/s41388-025-03661-0","DOIUrl":"10.1038/s41388-025-03661-0","url":null,"abstract":"RNA modification recognition proteins are crucial in cancer development and progression. Among all RNA modification-related proteins (RMRPs), Weighted Gene Co-expression Network Analysis (WGCNA), combined with comprehensive bioinformatic analysis, suggests that NAT10—the sole known writer of N4-acetylcytidine (ac4C)—is a critical regulatory protein in colorectal cancer (CRC) progression. NAT10 facilitates the malignancy phenotypes and DNA damage repair in CRC cells via its ac4C transferase activity and regulation of PPAN. Specifically, NAT10 enhances the translation efficiency of PPAN via acetylation at the C744 and C747 sites. In addition, NAT10 promotes the translation of ac4C-modified MYC mRNA. MYC protein then enhances PPAN transcription through binding to the PPAN promoter. The newly identified ac4C reader protein MYBBP1A mediates NAT10-induced translation of both PPAN and MYC. We further found that VDR binds to the NAT10 promoter to activate its transcription, resulting in the high expression of NAT10 in CRC. Xenograft studies and clinical data confirmed the role of the NAT10-PPAN axis in promoting CRC development and DNA damage repair. Collectively, our study reveals the role and underlying mechanism of mRNA ac4C modification in CRC progression, providing critical potential targets for CRC drug development.","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":"45 4","pages":"558-576"},"PeriodicalIF":7.3,"publicationDate":"2025-12-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145846820","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}
引用次数: 0
LAMP1 enhances DNA-PKcs-mediated AKT phosphorylation and VEGFC secretion to promote lymph node metastasis in esophageal squamous cell carcinoma LAMP1增强dna - pkcs介导的AKT磷酸化和VEGFC分泌促进食管鳞状细胞癌淋巴结转移。
IF 7.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-20 DOI: 10.1038/s41388-025-03664-x
Yikang Wang, Chang Yuan, Zhichao Liu, Ning Ma, Cong Qi, Boyao Yu, Haizhen Jin, Chenyang Tian, Chunguang Li, Zhigang Li
Lymph node metastasis (LNM) is a major determinant of poor prognosis in esophageal squamous cell carcinoma (ESCC), with patients remaining at high risk of postoperative recurrence in the absence of reliable predictors or effective interventions. To address this unmet clinical need, we perform proteomic and bioinformatic analyses and identify lysosome-associated membrane protein 1 (LAMP1) as a critical driver of LNM in ESCC. LAMP1 is markedly overexpressed in primary tumors that with LNM and correlates with worse survival outcomes across independent cohorts. Gain- and loss-of-function experiments demonstrate that LAMP1 promotes lymphangiogenesis and lymphatic metastasis both in vitro and in vivo. Mechanistically, LAMP1 interacts with DNA-PKcs, mediates AKT activation, leading to VEGFC upregulation and secretion, thereby driving lymphangiogenesis and metastasis. Importantly, treatment with the LAMP1 inhibitor Parishin C significantly suppresses LNM in preclinical ESCC models. These findings uncover a previously unrecognized LAMP1/DNA-PKcs/AKT/VEGFC signaling axis that promotes ESCC progression and highlight LAMP1 as a promising biomarker and therapeutic target for preventing LNM and improving patient outcomes.
淋巴结转移(LNM)是食管鳞状细胞癌(ESCC)预后不良的主要决定因素,在缺乏可靠的预测因素或有效干预措施的情况下,患者术后复发的风险仍然很高。为了解决这一未满足的临床需求,我们进行了蛋白质组学和生物信息学分析,并确定溶酶体相关膜蛋白1 (LAMP1)是ESCC中LNM的关键驱动因素。LAMP1在合并LNM的原发肿瘤中明显过表达,并且在独立队列中与较差的生存结果相关。获得和丧失功能的实验表明,LAMP1在体外和体内都促进淋巴管生成和淋巴转移。在机制上,LAMP1与DNA-PKcs相互作用,介导AKT活化,导致VEGFC上调和分泌,从而驱动淋巴管生成和转移。重要的是,使用LAMP1抑制剂Parishin C治疗可显著抑制临床前ESCC模型中的LNM。这些发现揭示了先前未被识别的LAMP1/DNA-PKcs/AKT/VEGFC信号轴促进ESCC进展,并突出了LAMP1作为预防LNM和改善患者预后的有前途的生物标志物和治疗靶点。
{"title":"LAMP1 enhances DNA-PKcs-mediated AKT phosphorylation and VEGFC secretion to promote lymph node metastasis in esophageal squamous cell carcinoma","authors":"Yikang Wang,&nbsp;Chang Yuan,&nbsp;Zhichao Liu,&nbsp;Ning Ma,&nbsp;Cong Qi,&nbsp;Boyao Yu,&nbsp;Haizhen Jin,&nbsp;Chenyang Tian,&nbsp;Chunguang Li,&nbsp;Zhigang Li","doi":"10.1038/s41388-025-03664-x","DOIUrl":"10.1038/s41388-025-03664-x","url":null,"abstract":"Lymph node metastasis (LNM) is a major determinant of poor prognosis in esophageal squamous cell carcinoma (ESCC), with patients remaining at high risk of postoperative recurrence in the absence of reliable predictors or effective interventions. To address this unmet clinical need, we perform proteomic and bioinformatic analyses and identify lysosome-associated membrane protein 1 (LAMP1) as a critical driver of LNM in ESCC. LAMP1 is markedly overexpressed in primary tumors that with LNM and correlates with worse survival outcomes across independent cohorts. Gain- and loss-of-function experiments demonstrate that LAMP1 promotes lymphangiogenesis and lymphatic metastasis both in vitro and in vivo. Mechanistically, LAMP1 interacts with DNA-PKcs, mediates AKT activation, leading to VEGFC upregulation and secretion, thereby driving lymphangiogenesis and metastasis. Importantly, treatment with the LAMP1 inhibitor Parishin C significantly suppresses LNM in preclinical ESCC models. These findings uncover a previously unrecognized LAMP1/DNA-PKcs/AKT/VEGFC signaling axis that promotes ESCC progression and highlight LAMP1 as a promising biomarker and therapeutic target for preventing LNM and improving patient outcomes.","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":"45 4","pages":"534-548"},"PeriodicalIF":7.3,"publicationDate":"2025-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145800090","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}
引用次数: 0
Chromosome centromere copy number amplification associated with exceptional response in HER2-positive metastatic breast cancer patients 染色体着丝粒拷贝数扩增与her2阳性转移性乳腺癌患者的异常反应相关。
IF 7.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-20 DOI: 10.1038/s41388-025-03667-8
Charlotte Andrieu, Jojanneke Stoof, Dalal AlSultan, Laura Ivers, Jose Javier Berenguer Pina, Darko Skrobo, Jo Ballot, Debbie O’Reilly, Denis M. Collins, Alex J. Eustace, Cecily Quinn, Janice M. Walshe, Giuseppe Gullo, Naomi Walsh, John Crown
Metastatic breast cancer (MBC) is generally an incurable neoplasm. A small cohort of patients with HER2-positive MBC, however, achieve such prolonged remission without relapse following anti-HER2 therapy and chemotherapy, that it is speculated they might be cured. The genomes of these patients might provide insights into the underlying mechanisms for their successful treatment. Here, a total of 243 HER2-positive patients diagnosed with MBC between 2000 and 2015 were studied. Of these, 29 patients were identified as exceptional responders (ExR) with an overall survival (OS) > 60 months and no evidence of relapse, 54 patients with an OS > 60 months but who relapsed or developed progressive disease were defined as exceptional survivors (ExS), and 160 patients with an OS < 60 months were identified as short-term responders (STR). Whole-Genome Sequencing and centromere copy number (CCN) analysis was performed on 27 patients (12 ExR; 4 ExS; 11 STR). A significant amplification was observed in the centromeric regions of ExR, exhibiting higher CCN compared to the ExS and STR. Digital PCR validation of chromosome 4 centromere region D4Z1 copy number was not associated with ExR OS. Our results suggest that the amplification of centromere regions are associated with very prolonged remission and survival in patients with HER2-positive MBC.
转移性乳腺癌(MBC)通常是一种无法治愈的肿瘤。然而,一小部分her2阳性MBC患者在抗her2治疗和化疗后获得了如此长时间的缓解而没有复发,推测他们可能被治愈。这些患者的基因组可能为他们成功治疗的潜在机制提供见解。本研究共研究了2000年至2015年间诊断为MBC的243例her2阳性患者。其中,29例患者被确定为特殊应答者(ExR),总生存期(OS)为60个月,无复发迹象,54例患者总生存期(OS)为60个月,但复发或发展为进展性疾病,被定义为特殊幸存者(ExS), 160例患者有OS
{"title":"Chromosome centromere copy number amplification associated with exceptional response in HER2-positive metastatic breast cancer patients","authors":"Charlotte Andrieu,&nbsp;Jojanneke Stoof,&nbsp;Dalal AlSultan,&nbsp;Laura Ivers,&nbsp;Jose Javier Berenguer Pina,&nbsp;Darko Skrobo,&nbsp;Jo Ballot,&nbsp;Debbie O’Reilly,&nbsp;Denis M. Collins,&nbsp;Alex J. Eustace,&nbsp;Cecily Quinn,&nbsp;Janice M. Walshe,&nbsp;Giuseppe Gullo,&nbsp;Naomi Walsh,&nbsp;John Crown","doi":"10.1038/s41388-025-03667-8","DOIUrl":"10.1038/s41388-025-03667-8","url":null,"abstract":"Metastatic breast cancer (MBC) is generally an incurable neoplasm. A small cohort of patients with HER2-positive MBC, however, achieve such prolonged remission without relapse following anti-HER2 therapy and chemotherapy, that it is speculated they might be cured. The genomes of these patients might provide insights into the underlying mechanisms for their successful treatment. Here, a total of 243 HER2-positive patients diagnosed with MBC between 2000 and 2015 were studied. Of these, 29 patients were identified as exceptional responders (ExR) with an overall survival (OS) &gt; 60 months and no evidence of relapse, 54 patients with an OS &gt; 60 months but who relapsed or developed progressive disease were defined as exceptional survivors (ExS), and 160 patients with an OS &lt; 60 months were identified as short-term responders (STR). Whole-Genome Sequencing and centromere copy number (CCN) analysis was performed on 27 patients (12 ExR; 4 ExS; 11 STR). A significant amplification was observed in the centromeric regions of ExR, exhibiting higher CCN compared to the ExS and STR. Digital PCR validation of chromosome 4 centromere region D4Z1 copy number was not associated with ExR OS. Our results suggest that the amplification of centromere regions are associated with very prolonged remission and survival in patients with HER2-positive MBC.","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":"45 4","pages":"549-557"},"PeriodicalIF":7.3,"publicationDate":"2025-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.comhttps://www.nature.com/articles/s41388-025-03667-8.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145800087","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}
引用次数: 0
期刊
Oncogene
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