首页 > 最新文献

Cancer letters最新文献

英文 中文
Corrigendum to "Lycorine promotes IDH1 acetylation to induce mitochondrial dynamics imbalance in colorectal cancer cells" [Canc. Lett. 573 (2023) 216364].
IF 9.1 1区 医学 Q1 ONCOLOGY Pub Date : 2025-02-09 DOI: 10.1016/j.canlet.2025.217533
Fang-Fang Zhuo, Ling Li, Ting-Ting Liu, Xiao-Min Liang, Zhuo Yang, Yong-Zhe Zheng, Qian-Wei Luo, Jia-Hong Lu, Dan Liu, Ke-Wu Zeng, Peng-Fei Tu
{"title":"Corrigendum to \"Lycorine promotes IDH1 acetylation to induce mitochondrial dynamics imbalance in colorectal cancer cells\" [Canc. Lett. 573 (2023) 216364].","authors":"Fang-Fang Zhuo, Ling Li, Ting-Ting Liu, Xiao-Min Liang, Zhuo Yang, Yong-Zhe Zheng, Qian-Wei Luo, Jia-Hong Lu, Dan Liu, Ke-Wu Zeng, Peng-Fei Tu","doi":"10.1016/j.canlet.2025.217533","DOIUrl":"https://doi.org/10.1016/j.canlet.2025.217533","url":null,"abstract":"","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":" ","pages":"217533"},"PeriodicalIF":9.1,"publicationDate":"2025-02-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143390168","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
CircMFN2/miR-361-3p/ELK1 feedback loop promotes glutaminolysis and the progression of hepatocellular carcinoma.
IF 9.1 1区 医学 Q1 ONCOLOGY Pub Date : 2025-02-09 DOI: 10.1016/j.canlet.2025.217473
Xiaopei Hao, Xiangjun Qian, Chenxi Xie, Zhengzheng Wang, Xiaoqian Wang, Yang Ji, Xiaokai Zhang, Qingjun Li, Baishun Wan, Hong Cui, Li Wang, Nanmu Yang, Liang Qiao, Haibo Yu, Feng Han, Hao Zhuang, Jinxue Zhou

Current evidence indicates that circRNAs are involved in the development of multiple malignancies including hepatocellular carcinoma (HCC). However, the specific functions of circRNAs in HCC metabolism and progression and their underlying regulatory mechanisms remain unclear. We have identified a novel circRNA circMFN2, by bioinformatics analysis of circRNA microarray data from the GEO database. The levels of circMFN2 were assessed in HCC cell lines and tissues, and its clinical relevance was assessed. The effect of circMFN2 on HCC cells was evaluated in vitro and in vivo. The effect of ELK1 on glutaminolysis and HCC progression was also explored. Patients with HCC and high circMFN2 expression exhibited worse survival outcomes. Functionally, downregulation of circMFN2 repressed the proliferation, invasion, and migration of HCC cells in vitro, whereas ectopic expression of circMFN2 had the opposite effects. The effects of tumor enhancement by circMFN2 on HCC were confirmed by in vivo experiments. Mechanistically, circMFN2 acted as a sponge for miR-361-3p, leading to the upregulation of its target ELK1, whereas ELK1 was enriched in the MFN2 promoter to enhance the transcription and expression of MFN2, indirectly leading to the upregulation of circMFN2. Additionally, we found that circMFN2 promotes glutaminolysis in HCC by increasing ELK1 phosphorylation. We concluded that circMFN2 facilitates HCC progression via a circMFN2/miR-361-3p/ELK1 feedback loop, which promotes glutaminolysis mediated by the upregulation of phosphorylated ELK1. Therefore, circMFN2 not only serves as a potential prognostic indicator, but it could also serve as a therapeutic target for HCC. Further studies are warranted.

{"title":"CircMFN2/miR-361-3p/ELK1 feedback loop promotes glutaminolysis and the progression of hepatocellular carcinoma.","authors":"Xiaopei Hao, Xiangjun Qian, Chenxi Xie, Zhengzheng Wang, Xiaoqian Wang, Yang Ji, Xiaokai Zhang, Qingjun Li, Baishun Wan, Hong Cui, Li Wang, Nanmu Yang, Liang Qiao, Haibo Yu, Feng Han, Hao Zhuang, Jinxue Zhou","doi":"10.1016/j.canlet.2025.217473","DOIUrl":"https://doi.org/10.1016/j.canlet.2025.217473","url":null,"abstract":"<p><p>Current evidence indicates that circRNAs are involved in the development of multiple malignancies including hepatocellular carcinoma (HCC). However, the specific functions of circRNAs in HCC metabolism and progression and their underlying regulatory mechanisms remain unclear. We have identified a novel circRNA circMFN2, by bioinformatics analysis of circRNA microarray data from the GEO database. The levels of circMFN2 were assessed in HCC cell lines and tissues, and its clinical relevance was assessed. The effect of circMFN2 on HCC cells was evaluated in vitro and in vivo. The effect of ELK1 on glutaminolysis and HCC progression was also explored. Patients with HCC and high circMFN2 expression exhibited worse survival outcomes. Functionally, downregulation of circMFN2 repressed the proliferation, invasion, and migration of HCC cells in vitro, whereas ectopic expression of circMFN2 had the opposite effects. The effects of tumor enhancement by circMFN2 on HCC were confirmed by in vivo experiments. Mechanistically, circMFN2 acted as a sponge for miR-361-3p, leading to the upregulation of its target ELK1, whereas ELK1 was enriched in the MFN2 promoter to enhance the transcription and expression of MFN2, indirectly leading to the upregulation of circMFN2. Additionally, we found that circMFN2 promotes glutaminolysis in HCC by increasing ELK1 phosphorylation. We concluded that circMFN2 facilitates HCC progression via a circMFN2/miR-361-3p/ELK1 feedback loop, which promotes glutaminolysis mediated by the upregulation of phosphorylated ELK1. Therefore, circMFN2 not only serves as a potential prognostic indicator, but it could also serve as a therapeutic target for HCC. Further studies are warranted.</p>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":" ","pages":"217473"},"PeriodicalIF":9.1,"publicationDate":"2025-02-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143398197","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
Traditional medicine Bazi Bushen potentiates immunosurveillance of senescent liver cancer cells via cGAS-STING signaling activation in macrophages. 传统药物巴子布申能通过激活巨噬细胞中的cGAS-STING信号增强对衰老肝癌细胞的免疫监视。
IF 9.1 1区 医学 Q1 ONCOLOGY Pub Date : 2025-02-08 DOI: 10.1016/j.canlet.2025.217544
Fuxue Xing, Hongwei Lv, Wei Xiang, Liang Wang, Qianni Zong, Guishuai Lv, Chunying Liu, Qiyu Feng, Hongyang Wang, Wen Yang

Senescent cancer cells often evade immune clearance to exert profound effects on cancer progression and therapy resistance. Improving immunosurveillance to eliminate senescent cancer cells is a crucial measure to enhance anti-cancer therapy. Bazi Bushen (BZBS) is a traditional medicine with the function of relieving fatigue and delaying ageing, but its role in tumor treatment remains poorly understood. Herein, we find that BZBS promotes immunosurveillance of both chemotherapy- and oncogene-induced senescent liver cancer cells, further leading to enhanced chemotherapy efficacy and dramatic tumor repression in mice. Mechanistically, BZBS induces mitochondrial DNA leakage by mitochondrial damage to further activate cGAS-STING signaling in macrophages. Subsequently, cGAS-STING signaling activation in macrophages recruits CD8+ T cells into tumor and promotes the anti-tumor activity of CD8+ T cells to eradicate senescent cancer cells. Furthermore, host STING is responsible for BZBS-mediated immunosurveillance of senescent liver cancer cells in mice. Therefore, our findings unveil the role of traditional medicine BZBS in activating cGAS-STING signaling and potentiating senescence immunosurveillance to enhance anti-cancer therapy.

{"title":"Traditional medicine Bazi Bushen potentiates immunosurveillance of senescent liver cancer cells via cGAS-STING signaling activation in macrophages.","authors":"Fuxue Xing, Hongwei Lv, Wei Xiang, Liang Wang, Qianni Zong, Guishuai Lv, Chunying Liu, Qiyu Feng, Hongyang Wang, Wen Yang","doi":"10.1016/j.canlet.2025.217544","DOIUrl":"https://doi.org/10.1016/j.canlet.2025.217544","url":null,"abstract":"<p><p>Senescent cancer cells often evade immune clearance to exert profound effects on cancer progression and therapy resistance. Improving immunosurveillance to eliminate senescent cancer cells is a crucial measure to enhance anti-cancer therapy. Bazi Bushen (BZBS) is a traditional medicine with the function of relieving fatigue and delaying ageing, but its role in tumor treatment remains poorly understood. Herein, we find that BZBS promotes immunosurveillance of both chemotherapy- and oncogene-induced senescent liver cancer cells, further leading to enhanced chemotherapy efficacy and dramatic tumor repression in mice. Mechanistically, BZBS induces mitochondrial DNA leakage by mitochondrial damage to further activate cGAS-STING signaling in macrophages. Subsequently, cGAS-STING signaling activation in macrophages recruits CD8<sup>+</sup> T cells into tumor and promotes the anti-tumor activity of CD8<sup>+</sup> T cells to eradicate senescent cancer cells. Furthermore, host STING is responsible for BZBS-mediated immunosurveillance of senescent liver cancer cells in mice. Therefore, our findings unveil the role of traditional medicine BZBS in activating cGAS-STING signaling and potentiating senescence immunosurveillance to enhance anti-cancer therapy.</p>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":" ","pages":"217544"},"PeriodicalIF":9.1,"publicationDate":"2025-02-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143390171","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
A Novel TLR4 Accessory Molecule Drives Hepatic Oncogenesis through Tumor-Associated Macrophages.
IF 9.1 1区 医学 Q1 ONCOLOGY Pub Date : 2025-02-08 DOI: 10.1016/j.canlet.2025.217543
Doyeon Kim, Carter A Allen, Dongjun Chung, Lingbin Meng, Xiaoli Zhang, Wenqing Zhang, Yuli Ouyang, Zihai Li, Feng Hong

Tumor-associated macrophages (TAMs) play a crucial role in the tumor microenvironment, yet the roles and mechanisms of TAMs in inflammation-associated oncogenesis remain enigmatic. We report that protein canopy homolog 2 (CNPY2) functions as a novel TLR4 regulator, promoting cytokine production in macrophages. CNPY2 binds directly to TLR4. Cnpy2 deficiency reduces cell surface expression of TLR4, nuclear translocation of NFκB and cytokine production in macrophages. Macrophage-specific CNPY2 deficiency significantly decreases cytokine production in macrophages and reduces hepatocarcinogenesis in a diethylnitrosamine (DEN)-induced liver cancer model. RNA-sequencing analysis revealed Cnpy2 knockout decreased the mRNA level and cell surface expression of two VEGF receptors, Flt1 and Kdr, compared to those in WT counterparts, resulting in inhibition of macrophage tumor infiltration. Cnpy2 knockout inhibits NFκB2/p52-mediated transcription of Flt1 and Kdr in macrophages. These findings demonstrate that CNPY2 regulates macrophages in both inflammation and hepatocarcinogenesis and may serve as a therapeutic target for cancer.

{"title":"A Novel TLR4 Accessory Molecule Drives Hepatic Oncogenesis through Tumor-Associated Macrophages.","authors":"Doyeon Kim, Carter A Allen, Dongjun Chung, Lingbin Meng, Xiaoli Zhang, Wenqing Zhang, Yuli Ouyang, Zihai Li, Feng Hong","doi":"10.1016/j.canlet.2025.217543","DOIUrl":"https://doi.org/10.1016/j.canlet.2025.217543","url":null,"abstract":"<p><p>Tumor-associated macrophages (TAMs) play a crucial role in the tumor microenvironment, yet the roles and mechanisms of TAMs in inflammation-associated oncogenesis remain enigmatic. We report that protein canopy homolog 2 (CNPY2) functions as a novel TLR4 regulator, promoting cytokine production in macrophages. CNPY2 binds directly to TLR4. Cnpy2 deficiency reduces cell surface expression of TLR4, nuclear translocation of NFκB and cytokine production in macrophages. Macrophage-specific CNPY2 deficiency significantly decreases cytokine production in macrophages and reduces hepatocarcinogenesis in a diethylnitrosamine (DEN)-induced liver cancer model. RNA-sequencing analysis revealed Cnpy2 knockout decreased the mRNA level and cell surface expression of two VEGF receptors, Flt1 and Kdr, compared to those in WT counterparts, resulting in inhibition of macrophage tumor infiltration. Cnpy2 knockout inhibits NFκB2/p52-mediated transcription of Flt1 and Kdr in macrophages. These findings demonstrate that CNPY2 regulates macrophages in both inflammation and hepatocarcinogenesis and may serve as a therapeutic target for cancer.</p>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":" ","pages":"217543"},"PeriodicalIF":9.1,"publicationDate":"2025-02-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143390167","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
Unveiling FKBP7 as an Early Endoplasmic Reticulum Sentinel in Pancreatic Stellate Cell Activation, Collagen Remodelling and Tumor Progression.
IF 9.1 1区 医学 Q1 ONCOLOGY Pub Date : 2025-02-07 DOI: 10.1016/j.canlet.2025.217538
Christophe Quemerais, Christine Jean, Alexia Brunel, Emilie Decaup, Guillaume Labrousse, Hippolyte Audureau, Jérôme Raffenne, Ismahane Belhabib, Jérôme Cros, Aurélie Perraud, Nelson Dusetti, Remy Nicolle, Muriel Mathonnet, Stéphane Pyronnet, Yvan Martineau, Marjorie Fanjul, Corinne Bousquet

In pancreatic ductal adenocarcinoma (PDAC), fibroblast activation leads to excessive secretion of extracellular matrix (ECM) and soluble factors that regulate tumor progression, prompting investigation into endoplasmic reticulum (ER)-resident proteins that may support this activation. We identified FKBP7, a peptidyl-prolyl isomerase in the ER, as overexpressed in PDAC stroma compared to cancer cells, and in patients with favorable prognosis. Analysis of single-cell RNA sequencing databases revealed FKBP7 expression in pancreatic stellate cells (PSCs) and cancer-associated fibroblasts (CAFs). When analyzed by immunohistochemistry on PDAC patient tissues, FKBP7 emerged as an early activation marker in the preneoplastic stroma, preceding αSMA expression, and responding to FAK- and TGFβ-induced stiffening and pro-fibrotic programs in PSCs. Functional analyses revealed that FKBP7 knockdown in PSCs enhanced contractility, Rho/ FAK signaling, and secretion of pro-inflammatory cytokines as well as remodeling of type I collagen, promoting an activated phenotype and accelerating tumor growth in vivo. Conversely, FKBP7 expression supported a tumor-restraining (i.e. encapsulating) ECM characterized by type IV collagen. Mechanistically, FKBP7 interacts with BiP, and blocking this interaction instead leads to increased PSC secretion of type I collagen. Thus, FKBP7 serves as a novel PSC marker and ER regulator in a complex with BiP of the secretion of specific collagen subtypes, highlighting its potential to mediate ECM normalization and constrain PDAC tumorigenesis.

{"title":"Unveiling FKBP7 as an Early Endoplasmic Reticulum Sentinel in Pancreatic Stellate Cell Activation, Collagen Remodelling and Tumor Progression.","authors":"Christophe Quemerais, Christine Jean, Alexia Brunel, Emilie Decaup, Guillaume Labrousse, Hippolyte Audureau, Jérôme Raffenne, Ismahane Belhabib, Jérôme Cros, Aurélie Perraud, Nelson Dusetti, Remy Nicolle, Muriel Mathonnet, Stéphane Pyronnet, Yvan Martineau, Marjorie Fanjul, Corinne Bousquet","doi":"10.1016/j.canlet.2025.217538","DOIUrl":"https://doi.org/10.1016/j.canlet.2025.217538","url":null,"abstract":"<p><p>In pancreatic ductal adenocarcinoma (PDAC), fibroblast activation leads to excessive secretion of extracellular matrix (ECM) and soluble factors that regulate tumor progression, prompting investigation into endoplasmic reticulum (ER)-resident proteins that may support this activation. We identified FKBP7, a peptidyl-prolyl isomerase in the ER, as overexpressed in PDAC stroma compared to cancer cells, and in patients with favorable prognosis. Analysis of single-cell RNA sequencing databases revealed FKBP7 expression in pancreatic stellate cells (PSCs) and cancer-associated fibroblasts (CAFs). When analyzed by immunohistochemistry on PDAC patient tissues, FKBP7 emerged as an early activation marker in the preneoplastic stroma, preceding αSMA expression, and responding to FAK- and TGFβ-induced stiffening and pro-fibrotic programs in PSCs. Functional analyses revealed that FKBP7 knockdown in PSCs enhanced contractility, Rho/ FAK signaling, and secretion of pro-inflammatory cytokines as well as remodeling of type I collagen, promoting an activated phenotype and accelerating tumor growth in vivo. Conversely, FKBP7 expression supported a tumor-restraining (i.e. encapsulating) ECM characterized by type IV collagen. Mechanistically, FKBP7 interacts with BiP, and blocking this interaction instead leads to increased PSC secretion of type I collagen. Thus, FKBP7 serves as a novel PSC marker and ER regulator in a complex with BiP of the secretion of specific collagen subtypes, highlighting its potential to mediate ECM normalization and constrain PDAC tumorigenesis.</p>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":" ","pages":"217538"},"PeriodicalIF":9.1,"publicationDate":"2025-02-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143381688","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
YTHDF3 drives tumor growth and metastasis by recruiting eIF4B to promote Notch2 translation in breast cancer
IF 9.1 1区 医学 Q1 ONCOLOGY Pub Date : 2025-02-07 DOI: 10.1016/j.canlet.2025.217534
Hongyu Chen , Linling Lin , Zishan Qiao , Yifei Pei , Yiyang Gao , Kangliang Lou , Lulin Yang , Chengxi Li , Yueyang He , Jingwen Bai , Guojun Zhang
YTH domain family protein 3 (YTHDF3), an m6A RNA reader, is implicated in various cancers, but its role in breast cancer progression and metastasis remains unclear. In this study, we explore the oncogenic potential of YTHDF3 in breast cancer, focusing on its impact on epithelial-mesenchymal transition (EMT) and metastasis. We found that YTHDF3 is significantly upregulated in breast cancer tissues and associated with poor relapse-free survival (RFS). Functional studies demonstrated that YTHDF3 promotes EMT in breast cancer cell lines by enhancing cell migration, invasion, and metastasis in vivo. Mechanistically, we show that YTHDF3 regulates Notch2, a key driver of EMT, through an m6A-dependent mechanism. YTHDF3 binds to m6A-modified Notch2 mRNA and recruits eIF4B to facilitate its translation, leading to increased Notch2 translation and subsequent inducing EMT. Our findings highlight the importance of the YTHDF3-Notch2 axis in driving EMT and metastasis in breast cancer. Furthermore, targeting YTHDF3 with lipid nanoparticles (LNPs) encapsulating siRNA and indocyanine green (ICG) significantly suppressed tumor growth and lung metastasis while enabling real-time therapeutic monitoring via ICG fluorescence imaging. These findings establish YTHDF3 as a critical driver of EMT and metastasis through m6A-dependent Notch2 translation, highlighting its potential as a therapeutic target in breast cancer.
YTH结构域家族蛋白3(YTHDF3)是一种m6A RNA阅读器,与多种癌症有关联,但它在乳腺癌进展和转移中的作用仍不清楚。在本研究中,我们探讨了YTHDF3在乳腺癌中的致癌潜能,重点关注其对上皮-间质转化(EMT)和转移的影响。我们发现,YTHDF3在乳腺癌组织中显著上调,并与无复发生存率(RFS)低下有关。功能研究表明,YTHDF3 通过增强细胞迁移、侵袭和体内转移,促进乳腺癌细胞系的 EMT。从机理上讲,我们发现YTHDF3通过m6A依赖性机制调控EMT的关键驱动因子Notch2。YTHDF3与m6A修饰的Notch2 mRNA结合,并招募eIF4B以促进其翻译,从而导致Notch2翻译增加,进而诱导EMT。我们的研究结果凸显了YTHDF3-Notch2轴在驱动乳腺癌EMT和转移中的重要性。此外,用封装有 siRNA 和吲哚菁绿(ICG)的脂质纳米颗粒(LNPs)靶向 YTHDF3 能显著抑制肿瘤生长和肺转移,同时还能通过 ICG 荧光成像进行实时治疗监测。这些研究结果表明,YTHDF3是通过m6A依赖性Notch2翻译导致EMT和转移的关键驱动因素,突显了其作为乳腺癌治疗靶点的潜力。
{"title":"YTHDF3 drives tumor growth and metastasis by recruiting eIF4B to promote Notch2 translation in breast cancer","authors":"Hongyu Chen ,&nbsp;Linling Lin ,&nbsp;Zishan Qiao ,&nbsp;Yifei Pei ,&nbsp;Yiyang Gao ,&nbsp;Kangliang Lou ,&nbsp;Lulin Yang ,&nbsp;Chengxi Li ,&nbsp;Yueyang He ,&nbsp;Jingwen Bai ,&nbsp;Guojun Zhang","doi":"10.1016/j.canlet.2025.217534","DOIUrl":"10.1016/j.canlet.2025.217534","url":null,"abstract":"<div><div>YTH domain family protein 3 (YTHDF3), an m<sup>6</sup>A RNA reader, is implicated in various cancers, but its role in breast cancer progression and metastasis remains unclear. In this study, we explore the oncogenic potential of YTHDF3 in breast cancer, focusing on its impact on epithelial-mesenchymal transition (EMT) and metastasis. We found that YTHDF3 is significantly upregulated in breast cancer tissues and associated with poor relapse-free survival (RFS). Functional studies demonstrated that YTHDF3 promotes EMT in breast cancer cell lines by enhancing cell migration, invasion, and metastasis <em>in vivo</em>. Mechanistically, we show that YTHDF3 regulates Notch2, a key driver of EMT, through an m<sup>6</sup>A-dependent mechanism. YTHDF3 binds to m<sup>6</sup>A-modified Notch2 mRNA and recruits eIF4B to facilitate its translation, leading to increased Notch2 translation and subsequent inducing EMT. Our findings highlight the importance of the YTHDF3-Notch2 axis in driving EMT and metastasis in breast cancer. Furthermore, targeting YTHDF3 with lipid nanoparticles (LNPs) encapsulating siRNA and indocyanine green (ICG) significantly suppressed tumor growth and lung metastasis while enabling real-time therapeutic monitoring via ICG fluorescence imaging. These findings establish YTHDF3 as a critical driver of EMT and metastasis through m<sup>6</sup>A-dependent Notch2 translation, highlighting its potential as a therapeutic target in breast cancer.</div></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":"614 ","pages":"Article 217534"},"PeriodicalIF":9.1,"publicationDate":"2025-02-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143381689","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
Current status and prospects of targeted therapy for cholangiocarcinoma based on molecular characteristics
IF 9.1 1区 医学 Q1 ONCOLOGY Pub Date : 2025-02-07 DOI: 10.1016/j.canlet.2025.217540
Xiaowen Cui , Teng Huang , Tianyi Jiang , Hongyang Wang
Cholangiocarcinoma (CCA) is a serious public health issue due to its insidious onset and dismal prognosis. The past few years have witnessed and highlighted the development of understanding and management of CCA. The combination of gemcitabine and cisplatin (GP) chemotherapy regimen with immunotherapy using immune checkpoint inhibitors has been considered the new standard first-line treatment alternative for advanced CCA. Notably, the proportion of patients with advanced CCA with targetable genetic mutations is approximately 40 %, and these patients may be considered for molecularly targeted therapy in the second-line treatment. In this review, we highlight the advances and progress in targeted therapies for advanced CCA, with special attention to data from Asian populations, including Chinese. In addition, we present in detail the phosphatase tension homolog (PTEN), a novel biomarker for both of first-line chemotherapy and second-line targeted therapy in advanced CCA, and its ability to forecast prognosis in patients with CCA. The mechanisms of rapid resistance to targeted agents warrant further investigation and address in light of the development of new targeted therapies. Precision medicine is gradually playing an increasing role in achieving optimal therapeutic outcomes.
{"title":"Current status and prospects of targeted therapy for cholangiocarcinoma based on molecular characteristics","authors":"Xiaowen Cui ,&nbsp;Teng Huang ,&nbsp;Tianyi Jiang ,&nbsp;Hongyang Wang","doi":"10.1016/j.canlet.2025.217540","DOIUrl":"10.1016/j.canlet.2025.217540","url":null,"abstract":"<div><div>Cholangiocarcinoma (CCA) is a serious public health issue due to its insidious onset and dismal prognosis. The past few years have witnessed and highlighted the development of understanding and management of CCA. The combination of gemcitabine and cisplatin (GP) chemotherapy regimen with immunotherapy using immune checkpoint inhibitors has been considered the new standard first-line treatment alternative for advanced CCA. Notably, the proportion of patients with advanced CCA with targetable genetic mutations is approximately 40 %, and these patients may be considered for molecularly targeted therapy in the second-line treatment. In this review, we highlight the advances and progress in targeted therapies for advanced CCA, with special attention to data from Asian populations, including Chinese. In addition, we present in detail the phosphatase tension homolog (PTEN), a novel biomarker for both of first-line chemotherapy and second-line targeted therapy in advanced CCA, and its ability to forecast prognosis in patients with CCA. The mechanisms of rapid resistance to targeted agents warrant further investigation and address in light of the development of new targeted therapies. Precision medicine is gradually playing an increasing role in achieving optimal therapeutic outcomes.</div></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":"614 ","pages":"Article 217540"},"PeriodicalIF":9.1,"publicationDate":"2025-02-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143381498","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
SQLE amplification accelerates esophageal squamous cell carcinoma tumorigenesis and metastasis through oncometabolite 2,3-oxidosqualene repressing Hippo pathway.
IF 9.1 1区 医学 Q1 ONCOLOGY Pub Date : 2025-02-07 DOI: 10.1016/j.canlet.2025.217528
Xuesong Liu, Mengzhu Lv, Bicong Feng, Ying Gong, Qingjie Min, Yan Wang, Qingnan Wu, Jie Chen, Dongyu Zhao, Jinting Li, Weimin Zhang, Qimin Zhan

Esophageal squamous cell carcinoma (ESCC) is one of the most prevalent cancers worldwide, characterized by a dismal prognosis and elusive therapeutic targets. Dysregulated cholesterol metabolism is a critical hallmark of cancer cells, facilitating tumor progression. Here, using whole genome sequencing data from several ESCC cohorts, we identified the important role of squalene epoxidase (SQLE) in promoting ESCC tumorigenesis and metastasis. Specifically, our findings highlight the significance of 2,3-oxidosqualene, an intermediate metabolite of cholesterol biosynthesis, synthesized by SQLE and metabolized by lanosterol synthase (LSS), as a key regulator of ESCC progression. Mechanistically, the interaction between 2,3-oxidosqualene and vinculin enhances the nuclear accumulation of Yes-associated protein 1 (YAP), thereby increasing YAP/TEAD-dependent gene expression, and accelerating both tumor growth and metastasis. In a 4-nitroquinoline 1-oxide (4-NQO)-induced ESCC mouse model, overexpression of Sqle resulted in accelerated tumorigenesis compared to wild-type controls, highlighting the pivotal role of SQLE in vivo. Furthermore, elevated SQLE expression in ESCC patients correlates with poorer prognoses, suggesting potential therapeutic avenues for ESCC treatment. In conclusion, our study elucidates the oncogenic function of 2,3-oxidosqualene as a naturally occurring metabolite and proposes modulation of its levels as a promising therapeutic strategy for ESCC.

食管鳞状细胞癌(ESCC)是全球发病率最高的癌症之一,其特点是预后不良和治疗靶点难以捉摸。胆固醇代谢失调是癌细胞的一个重要特征,有助于肿瘤的进展。在这里,我们利用来自多个 ESCC 队列的全基因组测序数据,确定了角鲨烯环氧化物酶(SQLE)在促进 ESCC 肿瘤发生和转移中的重要作用。具体而言,我们的研究结果强调了2,3-氧化角鲨烯作为ESCC进展的关键调控因子的重要性,角鲨烯是胆固醇生物合成的中间代谢产物,由SQLE合成并由羊毛甾醇合成酶(LSS)代谢。从机理上讲,2,3-氧化角鲨烯和长春花苷之间的相互作用会增强Yes相关蛋白1(YAP)的核积累,从而增加YAP/TEAD依赖性基因的表达,加速肿瘤的生长和转移。在4-硝基喹啉-1-氧化物(4-NQO)诱导的ESCC小鼠模型中,与野生型对照组相比,Sqle的过表达导致肿瘤发生加速,这突出表明了SQLE在体内的关键作用。此外,SQLE 在 ESCC 患者中的高表达与较差的预后相关,这为 ESCC 的治疗提供了潜在的治疗途径。总之,我们的研究阐明了 2,3-氧化角鲨烯作为一种天然代谢物的致癌功能,并提出调节其水平是一种治疗 ESCC 的可行策略。
{"title":"SQLE amplification accelerates esophageal squamous cell carcinoma tumorigenesis and metastasis through oncometabolite 2,3-oxidosqualene repressing Hippo pathway.","authors":"Xuesong Liu, Mengzhu Lv, Bicong Feng, Ying Gong, Qingjie Min, Yan Wang, Qingnan Wu, Jie Chen, Dongyu Zhao, Jinting Li, Weimin Zhang, Qimin Zhan","doi":"10.1016/j.canlet.2025.217528","DOIUrl":"https://doi.org/10.1016/j.canlet.2025.217528","url":null,"abstract":"<p><p>Esophageal squamous cell carcinoma (ESCC) is one of the most prevalent cancers worldwide, characterized by a dismal prognosis and elusive therapeutic targets. Dysregulated cholesterol metabolism is a critical hallmark of cancer cells, facilitating tumor progression. Here, using whole genome sequencing data from several ESCC cohorts, we identified the important role of squalene epoxidase (SQLE) in promoting ESCC tumorigenesis and metastasis. Specifically, our findings highlight the significance of 2,3-oxidosqualene, an intermediate metabolite of cholesterol biosynthesis, synthesized by SQLE and metabolized by lanosterol synthase (LSS), as a key regulator of ESCC progression. Mechanistically, the interaction between 2,3-oxidosqualene and vinculin enhances the nuclear accumulation of Yes-associated protein 1 (YAP), thereby increasing YAP/TEAD-dependent gene expression, and accelerating both tumor growth and metastasis. In a 4-nitroquinoline 1-oxide (4-NQO)-induced ESCC mouse model, overexpression of Sqle resulted in accelerated tumorigenesis compared to wild-type controls, highlighting the pivotal role of SQLE in vivo. Furthermore, elevated SQLE expression in ESCC patients correlates with poorer prognoses, suggesting potential therapeutic avenues for ESCC treatment. In conclusion, our study elucidates the oncogenic function of 2,3-oxidosqualene as a naturally occurring metabolite and proposes modulation of its levels as a promising therapeutic strategy for ESCC.</p>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":" ","pages":"217528"},"PeriodicalIF":9.1,"publicationDate":"2025-02-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143381685","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
Lactoferrin-Encapsulated Dichloroacetophenone (DAP) nanoparticles enhance drug delivery and anti-tumor efficacy in prostate cancer.
IF 9.1 1区 医学 Q1 ONCOLOGY Pub Date : 2025-02-07 DOI: 10.1016/j.canlet.2025.217522
Sugarniya Subramaniam, Varinder Jeet, Jennifer H Gunter, Judith A Clements, Srilakshmi Srinivasan, Amirali Popat, Jyotsna Batra

Pyruvate Dehydrogenase Kinase 1 (PDK1) regulates glycolysis and oxidative phosphorylation pathways and is linked to prostate cancer metastasis and poor prognosis. The therapeutic application of 2,2-dichloroacetophenone (DAP), a PDK1 inhibitor, remains underexplored in prostate cancer. In this study we demonstrated that DAP exhibited a superior ability to inhibit prostate cancer cell proliferation, migration and colony formation at a lower concentration (20 μM) compared to a previously established inhibitor, dichloroacetate (DCA), which required concentrations of 30 mM or higher. However, poor aqueous solubility and lower stability of DAP limits its therapeutic application. Nano formulation of DAP with natural lactoferrin enhanced its dispersion and stability by increasing polydispersity index and intensity, and reduced zeta potential values upon conjugation that overcame the solubility limitations of DAP. The lactoferrin-DAP nanoparticles exhibited enhanced therapeutic efficacy by precisely targeting prostate cancer cells that express high lactoferrin receptors and high anti-tumor activity in vitro (at 1 μM) and in mouse prostate tumor xenografts (20 mg/kg). Mechanistically, these nanoparticles induce apoptosis in cancer cells by inducing caspase3/7 activity and disrupting the glycolytic and oxidative phosphorylation pathways. Moreover, lactoferrin-conjugated DAP nanoparticles suppressed the viability of docetaxel-resistant cells exhibiting a higher inhibitory efficacy compared to free DAP and DCA. Targeting PDK1 through lactoferrin-conjugated DAP nanoparticles represents a potent targeted therapeutic strategy for disrupting prostate tumor metabolism and offers promising implications for overcoming drug resistance.

{"title":"Lactoferrin-Encapsulated Dichloroacetophenone (DAP) nanoparticles enhance drug delivery and anti-tumor efficacy in prostate cancer.","authors":"Sugarniya Subramaniam, Varinder Jeet, Jennifer H Gunter, Judith A Clements, Srilakshmi Srinivasan, Amirali Popat, Jyotsna Batra","doi":"10.1016/j.canlet.2025.217522","DOIUrl":"https://doi.org/10.1016/j.canlet.2025.217522","url":null,"abstract":"<p><p>Pyruvate Dehydrogenase Kinase 1 (PDK1) regulates glycolysis and oxidative phosphorylation pathways and is linked to prostate cancer metastasis and poor prognosis. The therapeutic application of 2,2-dichloroacetophenone (DAP), a PDK1 inhibitor, remains underexplored in prostate cancer. In this study we demonstrated that DAP exhibited a superior ability to inhibit prostate cancer cell proliferation, migration and colony formation at a lower concentration (20 μM) compared to a previously established inhibitor, dichloroacetate (DCA), which required concentrations of 30 mM or higher. However, poor aqueous solubility and lower stability of DAP limits its therapeutic application. Nano formulation of DAP with natural lactoferrin enhanced its dispersion and stability by increasing polydispersity index and intensity, and reduced zeta potential values upon conjugation that overcame the solubility limitations of DAP. The lactoferrin-DAP nanoparticles exhibited enhanced therapeutic efficacy by precisely targeting prostate cancer cells that express high lactoferrin receptors and high anti-tumor activity in vitro (at 1 μM) and in mouse prostate tumor xenografts (20 mg/kg). Mechanistically, these nanoparticles induce apoptosis in cancer cells by inducing caspase3/7 activity and disrupting the glycolytic and oxidative phosphorylation pathways. Moreover, lactoferrin-conjugated DAP nanoparticles suppressed the viability of docetaxel-resistant cells exhibiting a higher inhibitory efficacy compared to free DAP and DCA. Targeting PDK1 through lactoferrin-conjugated DAP nanoparticles represents a potent targeted therapeutic strategy for disrupting prostate tumor metabolism and offers promising implications for overcoming drug resistance.</p>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":" ","pages":"217522"},"PeriodicalIF":9.1,"publicationDate":"2025-02-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143381515","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
Elevated levels of circulating microbial-associated uremic toxins are associated with metastatic duodenopancreatic neuroendocrine tumors in patients with Multiple Endocrine Neoplasia Type 1
IF 9.1 1区 医学 Q1 ONCOLOGY Pub Date : 2025-02-07 DOI: 10.1016/j.canlet.2025.217537
Riccardo Ballarò , Amanda R. Wasylishen , Carolina R.C. Pieterman , Courtney Olsen , Ehsan Irajizad , Ranran Wu , Hiroyuki Katayama , Huiling Liu , Yining Cai , Ricardo A. León-Letelier , Jennifer B. Dennison , Steven Waguespack , Kim-Anh Do , Sunita K. Agarwal , Mary Walter , James Welch , Lee Weinstein , Jenny E. Blau , Smita Jha , Naris Nilubol , Johannes F. Fahrmann
Metastatic duodenopancreatic neuroendocrine tumors (dpNETs) are the primary cause of mortality among patients with Multiple Endocrine Neoplasia Type 1 (MEN1). Emerging evidence implicates the microbiome and microbial-derived secreted factors in promoting cancer development and progression. In the current study, we report that the circulating microbial-associated uremic toxins trimethylamine N-oxide (TMAO), indoxyl sulfate (IS), cresol sulfate (CS), cresol glucuronide (CG), and phenol sulfate (PS) are elevated in MEN1 patients with metastatic dpNETs. Proteomic- and metabolomic-based analysis of resected dpNET tissues from MEN1 patients also revealed detectable levels of uremic toxins that positively correlated with peptide-based signatures corresponding to Fusobacterium nucleatum, Faecalibacterium prausnitzii, and Klebsiella pneumoniae and negatively correlated with Streptococcus pneumoniae and Streptococcus thermophilus. A microbial-associated uremic toxin panel (MUTP) was developed and, in an independent case-control validation cohort, the panel yielded an area under the receiver operating characteristic curve (AUC) of 0.94 (95 % CI: 0.85–1.00) with 67 % sensitivity at 95 % specificity for identifying MEN1 patients with metastatic dpNETS. Increases in circulating microbial-associated uremic toxins during early stages of neoplasia were also found to be associated with poor overall survival in an Men1fl/flPdx1-CreTg mouse model of MEN1 pancreatic NETs. Our findings suggest that microbial dysbiosis is associated with disease aggressiveness and that increases in circulating microbial-associated uremic toxins may be a prognostic indication for MEN1 individuals who are at risk of having metastatic dpNETs.
{"title":"Elevated levels of circulating microbial-associated uremic toxins are associated with metastatic duodenopancreatic neuroendocrine tumors in patients with Multiple Endocrine Neoplasia Type 1","authors":"Riccardo Ballarò ,&nbsp;Amanda R. Wasylishen ,&nbsp;Carolina R.C. Pieterman ,&nbsp;Courtney Olsen ,&nbsp;Ehsan Irajizad ,&nbsp;Ranran Wu ,&nbsp;Hiroyuki Katayama ,&nbsp;Huiling Liu ,&nbsp;Yining Cai ,&nbsp;Ricardo A. León-Letelier ,&nbsp;Jennifer B. Dennison ,&nbsp;Steven Waguespack ,&nbsp;Kim-Anh Do ,&nbsp;Sunita K. Agarwal ,&nbsp;Mary Walter ,&nbsp;James Welch ,&nbsp;Lee Weinstein ,&nbsp;Jenny E. Blau ,&nbsp;Smita Jha ,&nbsp;Naris Nilubol ,&nbsp;Johannes F. Fahrmann","doi":"10.1016/j.canlet.2025.217537","DOIUrl":"10.1016/j.canlet.2025.217537","url":null,"abstract":"<div><div>Metastatic duodenopancreatic neuroendocrine tumors (dpNETs) are the primary cause of mortality among patients with Multiple Endocrine Neoplasia Type 1 (MEN1). Emerging evidence implicates the microbiome and microbial-derived secreted factors in promoting cancer development and progression. In the current study, we report that the circulating microbial-associated uremic toxins trimethylamine N-oxide (TMAO), indoxyl sulfate (IS), cresol sulfate (CS), cresol glucuronide (CG), and phenol sulfate (PS) are elevated in MEN1 patients with metastatic dpNETs. Proteomic- and metabolomic-based analysis of resected dpNET tissues from MEN1 patients also revealed detectable levels of uremic toxins that positively correlated with peptide-based signatures corresponding to <em>Fusobacterium nucleatum</em>, <em>Faecalibacterium prausnitzii,</em> and <em>Klebsiella pneumoniae</em> and negatively correlated with <em>Streptococcus pneumoniae</em> and <em>Streptococcus thermophilus</em>. A microbial-associated uremic toxin panel (MUTP) was developed and, in an independent case-control validation cohort, the panel yielded an area under the receiver operating characteristic curve (AUC) of 0.94 (95 % CI: 0.85–1.00) with 67 % sensitivity at 95 % specificity for identifying MEN1 patients with metastatic dpNETS. Increases in circulating microbial-associated uremic toxins during early stages of neoplasia were also found to be associated with poor overall survival in an <em>Men1</em><sup><em>fl/fl</em></sup><em>Pdx1-Cre</em><sup><em>Tg</em></sup> mouse model of MEN1 pancreatic NETs. Our findings suggest that microbial dysbiosis is associated with disease aggressiveness and that increases in circulating microbial-associated uremic toxins may be a prognostic indication for MEN1 individuals who are at risk of having metastatic dpNETs.</div></div>","PeriodicalId":9506,"journal":{"name":"Cancer letters","volume":"614 ","pages":"Article 217537"},"PeriodicalIF":9.1,"publicationDate":"2025-02-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143381502","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
期刊
Cancer letters
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:481959085
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