首页 > 最新文献

Cancer Communications最新文献

英文 中文
Galectin 3-binding protein (LGALS3BP) depletion attenuates hepatic fibrosis by reducing transforming growth factor-β1 (TGF-β1) availability and inhibits hepatocarcinogenesis. 通过减少转化生长因子-β1(TGF-β1)的可用性和抑制肝癌的发生,消耗凝集素 3 结合蛋白(LGALS3BP)可减轻肝纤维化。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-07-28 DOI: 10.1002/cac2.12600
Dae-Hwan Kim, Minjeong Sung, Myong-Suk Park, Eun-Gene Sun, Sumin Yoon, Kyung Hyun Yoo, Kamalakannan Radhakrishnan, Sung Yun Jung, Woo-Kyun Bae, Sang-Hee Cho, Ik-Joo Chung

Background: Increased Galectin 3-binding protein (LGALS3BP) serum levels have been used to assess hepatic fibrosis stages and the severity of hepatocellular carcinoma (HCC). Considering the crucial role of transforming growth factor-β1 (TGF-β1) in the emergence of these diseases, the present study tested the hypothesis that LGALS3BP regulates the TGF-β1 signaling pathway.

Methods: The expression levels of LGALS3BP and TGFB1 were analyzed in patients with metabolic dysfunction-associated steatohepatitis (MASH) and HCC. Multiple omics techniques, such as RNA-sequencing, transposase-accessible chromatin-sequencing assay, and liquid chromatography-tandem mass spectrometry proteomics, were used to identify the regulatory mechanisms for the LGALS3BP-TGF-β1 axis. The effects of altered TGF-β1 signaling by LGALS3BP were investigated in conditional LGALS3BP-knockin and LGALS3BP-knockout mice.

Results: In patients with MASH and HCC, the levels of LGALS3BP and TGFB1 exhibited positive correlations. Stimulation of LGALS3BP by the inflammatory cytokine interferon α in HCC cells or ectopic overexpression of LGALS3BP in hepatocytes promoted the expression levels of TGFB1. Aggravated fibrosis was observed in the livers of hepatocyte-specific LGALS3BP-knockin mice, with increased TGFB1 levels. LGALS3BP directly bound to and assembled integrin αV, an integral mediator required for releasing active TGF-β1 from extracellular latent complex with the rearranged F-actin cytoskeleton. The released TGF-β1 activated JunB transcription factor, which in turn promoted the TGF-β1 positive feedback loop. LGALS3BP deletion in the hepatocytes downregulated TGF-β1 signaling and CCl4 induced fibrosis. Moreover, LGALS3BP depletion hindered hepatocarcinogenesis by limiting the availability of fibrogenic TGF-β1.

Conclusion: LGALS3BP plays a crucial role in hepatic fibrosis and carcinogenesis by controlling the TGF-β1 signaling pathway, making it a promising therapeutic target in TGF-β1-related diseases.

背景:Galectin 3结合蛋白(LGALS3BP)血清水平的升高已被用于评估肝纤维化阶段和肝细胞癌(HCC)的严重程度。考虑到转化生长因子-β1(TGF-β1)在这些疾病的发生中的关键作用,本研究对 LGALS3BP 调节 TGF-β1 信号通路的假设进行了检验:方法:分析了代谢功能障碍相关性脂肪性肝炎(MASH)和HCC患者中LGALS3BP和TGFB1的表达水平。研究人员采用RNA测序、转座酶可访问染色质测序分析和液相色谱-串联质谱蛋白质组学等多种omics技术来确定LGALS3BP-TGF-β1轴的调控机制。在条件性LGALS3BP-knockin和LGALS3BP-knockout小鼠中研究了LGALS3BP改变TGF-β1信号传导的影响:结果:在MASH和HCC患者中,LGALS3BP和TGFB1的水平呈正相关。HCC 细胞中的炎性细胞因子干扰素 α 对 LGALS3BP 的刺激或肝细胞中 LGALS3BP 的异位过表达促进了 TGFB1 的表达水平。在肝细胞特异性LGALS3BP敲除小鼠的肝脏中观察到纤维化加重,TGFB1水平升高。LGALS3BP直接与整合素αV结合并组装,整合素αV是将活性TGF-β1从细胞外潜伏复合物与重新排列的F-肌动蛋白细胞骨架中释放出来所需的整合介质。释放的 TGF-β1 激活了 JunB 转录因子,而 JunB 又促进了 TGF-β1 的正反馈循环。肝细胞中 LGALS3BP 的缺失下调了 TGF-β1 信号传导和 CCl4 诱导的纤维化。此外,LGALS3BP缺失通过限制纤维化TGF-β1的可用性,阻碍了肝癌的发生:结论:LGALS3BP通过控制TGF-β1信号通路,在肝纤维化和肝癌发生过程中发挥着重要作用,因此是治疗TGF-β1相关疾病的理想靶点。
{"title":"Galectin 3-binding protein (LGALS3BP) depletion attenuates hepatic fibrosis by reducing transforming growth factor-β1 (TGF-β1) availability and inhibits hepatocarcinogenesis.","authors":"Dae-Hwan Kim, Minjeong Sung, Myong-Suk Park, Eun-Gene Sun, Sumin Yoon, Kyung Hyun Yoo, Kamalakannan Radhakrishnan, Sung Yun Jung, Woo-Kyun Bae, Sang-Hee Cho, Ik-Joo Chung","doi":"10.1002/cac2.12600","DOIUrl":"https://doi.org/10.1002/cac2.12600","url":null,"abstract":"<p><strong>Background: </strong>Increased Galectin 3-binding protein (LGALS3BP) serum levels have been used to assess hepatic fibrosis stages and the severity of hepatocellular carcinoma (HCC). Considering the crucial role of transforming growth factor-β1 (TGF-β1) in the emergence of these diseases, the present study tested the hypothesis that LGALS3BP regulates the TGF-β1 signaling pathway.</p><p><strong>Methods: </strong>The expression levels of LGALS3BP and TGFB1 were analyzed in patients with metabolic dysfunction-associated steatohepatitis (MASH) and HCC. Multiple omics techniques, such as RNA-sequencing, transposase-accessible chromatin-sequencing assay, and liquid chromatography-tandem mass spectrometry proteomics, were used to identify the regulatory mechanisms for the LGALS3BP-TGF-β1 axis. The effects of altered TGF-β1 signaling by LGALS3BP were investigated in conditional LGALS3BP-knockin and LGALS3BP-knockout mice.</p><p><strong>Results: </strong>In patients with MASH and HCC, the levels of LGALS3BP and TGFB1 exhibited positive correlations. Stimulation of LGALS3BP by the inflammatory cytokine interferon α in HCC cells or ectopic overexpression of LGALS3BP in hepatocytes promoted the expression levels of TGFB1. Aggravated fibrosis was observed in the livers of hepatocyte-specific LGALS3BP-knockin mice, with increased TGFB1 levels. LGALS3BP directly bound to and assembled integrin αV, an integral mediator required for releasing active TGF-β1 from extracellular latent complex with the rearranged F-actin cytoskeleton. The released TGF-β1 activated JunB transcription factor, which in turn promoted the TGF-β1 positive feedback loop. LGALS3BP deletion in the hepatocytes downregulated TGF-β1 signaling and CCl<sub>4</sub> induced fibrosis. Moreover, LGALS3BP depletion hindered hepatocarcinogenesis by limiting the availability of fibrogenic TGF-β1.</p><p><strong>Conclusion: </strong>LGALS3BP plays a crucial role in hepatic fibrosis and carcinogenesis by controlling the TGF-β1 signaling pathway, making it a promising therapeutic target in TGF-β1-related diseases.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-07-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141787312","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
Dysfunction of dendritic cells in tumor microenvironment and immunotherapy. 树突状细胞在肿瘤微环境和免疫疗法中的功能障碍。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-07-25 DOI: 10.1002/cac2.12596
Jie Chen, Yuhang Duan, Junye Che, Jianwei Zhu

Dendritic cells (DCs) comprise diverse cell populations that play critical roles in antigen presentation and triggering immune responses in the body. However, several factors impair the immune function of DCs and may promote immune evasion in cancer. Understanding the mechanism of DC dysfunction and the diverse functions of heterogeneous DCs in the tumor microenvironment (TME) is critical for designing effective strategies for cancer immunotherapy. Clinical applications targeting DCs summarized in this report aim to improve immune infiltration and enhance the biological function of DCs to modulate the TME to prevent cancer cells from evading the immune system. Herein, factors in the TME that induce DC dysfunction, such as cytokines, hypoxic environment, tumor exosomes and metabolites, and co-inhibitory molecules, have been described. Furthermore, several key signaling pathways involved in DC dysfunction and signal-relevant drugs evaluated in clinical trials were identified. Finally, this review provides an overview of current clinical immunotherapies targeting DCs, especially therapies with proven clinical outcomes, and explores future developments in DC immunotherapies.

树突状细胞(DC)由多种细胞群组成,在抗原呈递和引发体内免疫反应方面发挥着关键作用。然而,有几种因素会损害树突状细胞的免疫功能,并可能促进癌症的免疫逃避。了解肿瘤微环境(TME)中直流细胞功能障碍的机制和异质性直流细胞的多种功能对于设计有效的癌症免疫疗法至关重要。本报告总结的以直流电为靶点的临床应用旨在改善免疫浸润,增强直流电的生物功能,从而调节肿瘤微环境,防止癌细胞逃避免疫系统。本文描述了TME中诱导DC功能障碍的因素,如细胞因子、缺氧环境、肿瘤外泌体和代谢产物以及协同抑制分子。此外,还确定了参与直流电功能障碍的几种关键信号通路以及在临床试验中评估的信号相关药物。最后,本综述概述了目前针对直流电的临床免疫疗法,特别是已证实具有临床疗效的疗法,并探讨了直流电免疫疗法的未来发展。
{"title":"Dysfunction of dendritic cells in tumor microenvironment and immunotherapy.","authors":"Jie Chen, Yuhang Duan, Junye Che, Jianwei Zhu","doi":"10.1002/cac2.12596","DOIUrl":"https://doi.org/10.1002/cac2.12596","url":null,"abstract":"<p><p>Dendritic cells (DCs) comprise diverse cell populations that play critical roles in antigen presentation and triggering immune responses in the body. However, several factors impair the immune function of DCs and may promote immune evasion in cancer. Understanding the mechanism of DC dysfunction and the diverse functions of heterogeneous DCs in the tumor microenvironment (TME) is critical for designing effective strategies for cancer immunotherapy. Clinical applications targeting DCs summarized in this report aim to improve immune infiltration and enhance the biological function of DCs to modulate the TME to prevent cancer cells from evading the immune system. Herein, factors in the TME that induce DC dysfunction, such as cytokines, hypoxic environment, tumor exosomes and metabolites, and co-inhibitory molecules, have been described. Furthermore, several key signaling pathways involved in DC dysfunction and signal-relevant drugs evaluated in clinical trials were identified. Finally, this review provides an overview of current clinical immunotherapies targeting DCs, especially therapies with proven clinical outcomes, and explores future developments in DC immunotherapies.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141757320","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
Efficacy, safety and biomarkers of SG001 for patients with previously treated recurrent or metastatic cervical cancer: an open-label, multicenter, phase Ib trial. SG001对既往接受过治疗的复发性或转移性宫颈癌患者的疗效、安全性和生物标志物:一项开放标签、多中心、Ib期试验。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-07-23 DOI: 10.1002/cac2.12578
Jing Zuo, Wei Duan, Mingxuan Zhao, Zhendong Chen, Jie Lin, Huaqiu Shi, Ou Jiang, Youzhong Zhang, Meiyu Fang, Li Wang, Wei Wang, Yong Huang, Junyan Yu, Xiaoxue Zhang, Weiqing Pu, Deshun Hao, Fenglin She, Xiugao Yang, Ying Chen, Qizhi Tang, Xiao Zhang, Miao Niu, Yan'e Song, Lingying Wu
{"title":"Efficacy, safety and biomarkers of SG001 for patients with previously treated recurrent or metastatic cervical cancer: an open-label, multicenter, phase Ib trial.","authors":"Jing Zuo, Wei Duan, Mingxuan Zhao, Zhendong Chen, Jie Lin, Huaqiu Shi, Ou Jiang, Youzhong Zhang, Meiyu Fang, Li Wang, Wei Wang, Yong Huang, Junyan Yu, Xiaoxue Zhang, Weiqing Pu, Deshun Hao, Fenglin She, Xiugao Yang, Ying Chen, Qizhi Tang, Xiao Zhang, Miao Niu, Yan'e Song, Lingying Wu","doi":"10.1002/cac2.12578","DOIUrl":"https://doi.org/10.1002/cac2.12578","url":null,"abstract":"","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141751183","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 N4-acetylcytidine suppresses hepatocellular carcinoma progression by repressing eEF2-mediated HMGB2 mRNA translation. 通过抑制 eEF2 介导的 HMGB2 mRNA 翻译,靶向 N4-乙酰胞嘧啶可抑制肝细胞癌的进展。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-07-19 DOI: 10.1002/cac2.12595
Hailing Liu, Lei Xu, Shiwei Yue, Hongfei Su, Xing Chen, Qiumeng Liu, Hui Li, Huifang Liang, Xiaoping Chen, Jiefeng He, Zeyang Ding, Bixiang Zhang

Background: N4-acetylcytidine (ac4C) represents a novel messenger RNA (mRNA) modification, and its associated acetyltransferase N-acetyltransferase 10 (NAT10) plays a crucial role in the initiation and progression of tumors by regulating mRNA functionality. However, its role in hepatocellular carcinoma (HCC) development and prognosis is largely unknown. This study aimed to elucidate the role of NAT10-mediated ac4C in HCC progression and provide a promising therapeutic approach.

Methods: The ac4C levels were evaluated by dot blot and ultra-performance liquid chromatography-tandem mass spectrometry with harvested HCC tissues. The expression of NAT10 was investigated using quantitative real-time polymerase chain reaction, western blotting, and immunohistochemical staining across 91 cohorts of HCC patients. To explore the underlying mechanisms of NAT10-ac4C in HCC, we employed a comprehensive approach integrating acetylated RNA immunoprecipitation and sequencing, RNA sequencing and ribosome profiling analyses, along with RNA immunoprecipitation, RNA pull-down, mass spectrometry, and site-specific mutation analyses. The drug affinity responsive targets stability, cellular thermal shift assay, and surface plasmon resonance assays were performed to assess the specific binding of NAT10 and Panobinostat. Furthermore, the efficacy of targeting NAT10-ac4C for HCC treatment was elucidated through in vitro experiments using HCC cells and in vivo HCC mouse models.

Results: Our investigation revealed a significant increase in both the ac4C RNA level and NAT10 expression in HCC. Notably, elevated NAT10 expression was associated with poor outcomes in HCC patients. Functionally, silencing NAT10 suppressed HCC proliferation and metastasis in vitro and in vivo. Mechanistically, NAT10 stimulates the ac4C modification within the coding sequence (CDS) of high mobility group protein B2 (HMGB2), which subsequently enhances HMGB2 translation by facilitating eukaryotic elongation factor 2 (eEF2) binding to the ac4C sites on HMGB2 mRNA's CDS. Additionally, high-throughput compound library screening revealed Panobinostat as a potent inhibitor of NAT10-mediated ac4C modification. This inhibition significantly attenuated HCC growth and metastasis in both in vitro experiments using HCC cells and in vivo HCC mouse models.

Conclusions: Our study identified a novel oncogenic epi-transcriptome axis involving NAT10-ac4C/eEF2-HMGB2, which plays a pivotal role in regulating HCC growth and metastasis. The drug Panobinostat validates the therapeutic potential of targeting this axis for HCC treatment.

背景:N4-乙酰胞苷(ac4C)是一种新型的信使 RNA(mRNA)修饰,与之相关的乙酰转移酶 N-acetyltransferase 10(NAT10)通过调节 mRNA 的功能在肿瘤的发生和发展过程中起着至关重要的作用。然而,它在肝细胞癌(HCC)的发展和预后中的作用在很大程度上是未知的。本研究旨在阐明 NAT10 介导的 ac4C 在 HCC 进展中的作用,并提供一种有前景的治疗方法:方法:通过点印迹和超高效液相色谱-串联质谱法对采集的 HCC 组织中的 ac4C 水平进行评估。采用定量实时聚合酶链式反应、Western 印迹法和免疫组化染色法研究了 91 组 HCC 患者中 NAT10 的表达情况。为了探索 NAT10-ac4C 在 HCC 中的潜在机制,我们采用了一种综合方法,将乙酰化 RNA 免疫沉淀和测序、RNA 测序和核糖体图谱分析以及 RNA 免疫沉淀、RNA 拉取、质谱分析和位点特异性突变分析结合在一起。为了评估 NAT10 和 Panobinostat 的特异性结合,还进行了药物亲和力反应靶点稳定性、细胞热转移试验和表面等离子体共振试验。此外,通过使用 HCC 细胞的体外实验和体内 HCC 小鼠模型,阐明了靶向 NAT10-ac4C 治疗 HCC 的疗效:结果:我们的研究发现,HCC中的ac4C RNA水平和NAT10表达均有明显增加。值得注意的是,NAT10 表达的升高与 HCC 患者的不良预后有关。从功能上讲,沉默 NAT10 可抑制 HCC 在体外和体内的增殖和转移。从机理上讲,NAT10能刺激高迁移率基团蛋白B2(HMGB2)编码序列(CDS)中的ac4C修饰,随后通过促进真核延伸因子2(eEF2)与HMGB2 mRNA的CDS上的ac4C位点结合来增强HMGB2的翻译。此外,高通量化合物库筛选发现,Panobinostat 是 NAT10 介导的 ac4C 修饰的强效抑制剂。在使用 HCC 细胞进行的体外实验和体内 HCC 小鼠模型中,这种抑制都能明显减轻 HCC 的生长和转移:我们的研究发现了一个涉及 NAT10-ac4C/eEF2-HMGB2 的新型致癌外转录组轴,它在调控 HCC 生长和转移方面发挥着关键作用。药物 Panobinostat 验证了针对该轴治疗 HCC 的治疗潜力。
{"title":"Targeting N4-acetylcytidine suppresses hepatocellular carcinoma progression by repressing eEF2-mediated HMGB2 mRNA translation.","authors":"Hailing Liu, Lei Xu, Shiwei Yue, Hongfei Su, Xing Chen, Qiumeng Liu, Hui Li, Huifang Liang, Xiaoping Chen, Jiefeng He, Zeyang Ding, Bixiang Zhang","doi":"10.1002/cac2.12595","DOIUrl":"https://doi.org/10.1002/cac2.12595","url":null,"abstract":"<p><strong>Background: </strong>N4-acetylcytidine (ac4C) represents a novel messenger RNA (mRNA) modification, and its associated acetyltransferase N-acetyltransferase 10 (NAT10) plays a crucial role in the initiation and progression of tumors by regulating mRNA functionality. However, its role in hepatocellular carcinoma (HCC) development and prognosis is largely unknown. This study aimed to elucidate the role of NAT10-mediated ac4C in HCC progression and provide a promising therapeutic approach.</p><p><strong>Methods: </strong>The ac4C levels were evaluated by dot blot and ultra-performance liquid chromatography-tandem mass spectrometry with harvested HCC tissues. The expression of NAT10 was investigated using quantitative real-time polymerase chain reaction, western blotting, and immunohistochemical staining across 91 cohorts of HCC patients. To explore the underlying mechanisms of NAT10-ac4C in HCC, we employed a comprehensive approach integrating acetylated RNA immunoprecipitation and sequencing, RNA sequencing and ribosome profiling analyses, along with RNA immunoprecipitation, RNA pull-down, mass spectrometry, and site-specific mutation analyses. The drug affinity responsive targets stability, cellular thermal shift assay, and surface plasmon resonance assays were performed to assess the specific binding of NAT10 and Panobinostat. Furthermore, the efficacy of targeting NAT10-ac4C for HCC treatment was elucidated through in vitro experiments using HCC cells and in vivo HCC mouse models.</p><p><strong>Results: </strong>Our investigation revealed a significant increase in both the ac4C RNA level and NAT10 expression in HCC. Notably, elevated NAT10 expression was associated with poor outcomes in HCC patients. Functionally, silencing NAT10 suppressed HCC proliferation and metastasis in vitro and in vivo. Mechanistically, NAT10 stimulates the ac4C modification within the coding sequence (CDS) of high mobility group protein B2 (HMGB2), which subsequently enhances HMGB2 translation by facilitating eukaryotic elongation factor 2 (eEF2) binding to the ac4C sites on HMGB2 mRNA's CDS. Additionally, high-throughput compound library screening revealed Panobinostat as a potent inhibitor of NAT10-mediated ac4C modification. This inhibition significantly attenuated HCC growth and metastasis in both in vitro experiments using HCC cells and in vivo HCC mouse models.</p><p><strong>Conclusions: </strong>Our study identified a novel oncogenic epi-transcriptome axis involving NAT10-ac4C/eEF2-HMGB2, which plays a pivotal role in regulating HCC growth and metastasis. The drug Panobinostat validates the therapeutic potential of targeting this axis for HCC treatment.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141731006","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
Real-world data on ALK rearrangement test in Chinese advanced non-small cell lung cancer (RATICAL): a nationwide multicenter retrospective study. 中国晚期非小细胞肺癌ALK重排检测的真实世界数据(RATICAL):一项全国多中心回顾性研究。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-07-17 DOI: 10.1002/cac2.12593
Lin Li, Wencai Li, Chunyan Wu, Yanfeng Xi, Lei Guo, Yuan Ji, Lili Jiang, Ji Li, Jingping Yun, Gang Chen, Yuan Li, Yueping Liu, Dianbin Mu, Yuchen Han, Leina Sun, Qingxin Xia, Xiaodong Teng, Nanying Che, Wei Wu, Xueshan Qiu, Chao Liu, Xiaochu Yan, Daiqiang Li, Zhihong Zhang, Zhe Wang, Yujun Li, Zheng Wang, Lingchuan Guo, Xiu Nie, Jingshu Geng, Jianhua Zhou, Jianming Ying

Background: Anaplastic lymphoma kinase (ALK) test in advanced non-small cell lung cancer (NSCLC) can help physicians provide target therapies for patients harboring ALK gene rearrangement. This study aimed to investigate the real-world test patterns and positive rates of ALK gene rearrangements in advanced NSCLC.

Methods: In this real-world study (ChiCTR2000030266), patients with advanced NSCLC who underwent an ALK rearrangement test in 30 medical centers in China between October 1, 2018 and December 31, 2019 were retrospectively analyzed. Interpretation training was conducted before the study was initiated. Quality controls were performed at participating centers using immunohistochemistry (IHC)-VENTANA-D5F3. The positive ALK gene rearrangement rate and consistency rate were calculated. The associated clinicopathological characteristics of ALK gene rearrangement were investigated as well.

Results: The overall ALK gene rearrangement rate was 6.7% in 23,689 patients with advanced NSCLC and 8.2% in 17,436 patients with advanced lung adenocarcinoma. The quality control analysis of IHC-VENTANA-D5F3 revealed an intra-hospital consistency rate of 98.2% (879/895) and an inter-hospital consistency rate of 99.2% (646/651). IHC-VENTANA-D5F3 was used in 53.6%, real-time polymerase chain reaction (RT-PCR) in 25.4%, next-generation sequencing (NGS) in 18.3%, and fluorescence in-situ hybridization (FISH) in 15.9% in the adenocarcinoma subgroup. For specimens tested with multiple methods, the consistency rates confirmed by IHC-VENTANA-D5F3 were 98.0% (822/839) for FISH, 98.7% (1,222/1,238) for NGS, and 91.3% (146/160) for RT-PCR. The overall ALK gene rearrangement rates were higher in females, patients of ≤ 35 years old, never smokers, tumor cellularity of > 50, and metastatic specimens used for testing in the total NSCLC population and adenocarcinoma subgroup (all P < 0.05).

Conclusions: This study highlights the real-world variability and challenges of ALK test in advanced NSCLC, demonstrating a predominant use of IHC-VENTANA-D5F3 with high consistency and distinct clinicopathological features in ALK-positive patients. These findings underscore the need for a consensus on optimal test practices and support the development of refined ALK test strategies to enhance diagnostic accuracy and therapeutic decision-making in NSCLC.

背景:晚期非小细胞肺癌(NSCLC)中的无性淋巴瘤激酶(ALK)检测可帮助医生为携带ALK基因重排的患者提供靶向治疗。本研究旨在调查真实世界中晚期非小细胞肺癌 ALK 基因重排的检测模式和阳性率:在这项真实世界研究(ChiCTR2000030266)中,对2018年10月1日至2019年12月31日期间在中国30家医疗中心接受ALK基因重排检测的晚期NSCLC患者进行了回顾性分析。研究开始前进行了解读培训。参与中心使用免疫组化(IHC)-VENTANA-D5F3进行了质量控制。计算了ALK基因重排阳性率和一致性率。同时还调查了与ALK基因重排相关的临床病理特征:23689例晚期NSCLC患者的ALK基因重排率为6.7%,17436例晚期肺腺癌患者的ALK基因重排率为8.2%。IHC-VENTANA-D5F3的质控分析显示,医院内一致性率为98.2%(879/895),医院间一致性率为99.2%(646/651)。腺癌亚组中使用 IHC-VENTANA-D5F3 的占 53.6%,使用实时聚合酶链反应 (RT-PCR) 的占 25.4%,使用新一代测序 (NGS) 的占 18.3%,使用荧光原位杂交 (FISH) 的占 15.9%。对于采用多种方法检测的标本,经 IHC-VENTANA-D5F3 确认的一致性率分别为:FISH 98.0%(822/839),NGS 98.7%(1,222/1,238),RT-PCR 91.3%(146/160)。在所有NSCLC人群和腺癌亚组中,女性、年龄小于35岁、从不吸烟、肿瘤细胞度大于50、用于检测的转移标本的ALK基因重排率较高(均P<0.05):本研究强调了ALK检测在晚期NSCLC中的现实世界中的可变性和挑战,表明在ALK阳性患者中主要使用IHC-VENTANA-D5F3,一致性高,临床病理特征明显。这些发现强调了就最佳检测方法达成共识的必要性,并支持开发完善的ALK检测策略,以提高NSCLC的诊断准确性和治疗决策。
{"title":"Real-world data on ALK rearrangement test in Chinese advanced non-small cell lung cancer (RATICAL): a nationwide multicenter retrospective study.","authors":"Lin Li, Wencai Li, Chunyan Wu, Yanfeng Xi, Lei Guo, Yuan Ji, Lili Jiang, Ji Li, Jingping Yun, Gang Chen, Yuan Li, Yueping Liu, Dianbin Mu, Yuchen Han, Leina Sun, Qingxin Xia, Xiaodong Teng, Nanying Che, Wei Wu, Xueshan Qiu, Chao Liu, Xiaochu Yan, Daiqiang Li, Zhihong Zhang, Zhe Wang, Yujun Li, Zheng Wang, Lingchuan Guo, Xiu Nie, Jingshu Geng, Jianhua Zhou, Jianming Ying","doi":"10.1002/cac2.12593","DOIUrl":"https://doi.org/10.1002/cac2.12593","url":null,"abstract":"<p><strong>Background: </strong>Anaplastic lymphoma kinase (ALK) test in advanced non-small cell lung cancer (NSCLC) can help physicians provide target therapies for patients harboring ALK gene rearrangement. This study aimed to investigate the real-world test patterns and positive rates of ALK gene rearrangements in advanced NSCLC.</p><p><strong>Methods: </strong>In this real-world study (ChiCTR2000030266), patients with advanced NSCLC who underwent an ALK rearrangement test in 30 medical centers in China between October 1, 2018 and December 31, 2019 were retrospectively analyzed. Interpretation training was conducted before the study was initiated. Quality controls were performed at participating centers using immunohistochemistry (IHC)-VENTANA-D5F3. The positive ALK gene rearrangement rate and consistency rate were calculated. The associated clinicopathological characteristics of ALK gene rearrangement were investigated as well.</p><p><strong>Results: </strong>The overall ALK gene rearrangement rate was 6.7% in 23,689 patients with advanced NSCLC and 8.2% in 17,436 patients with advanced lung adenocarcinoma. The quality control analysis of IHC-VENTANA-D5F3 revealed an intra-hospital consistency rate of 98.2% (879/895) and an inter-hospital consistency rate of 99.2% (646/651). IHC-VENTANA-D5F3 was used in 53.6%, real-time polymerase chain reaction (RT-PCR) in 25.4%, next-generation sequencing (NGS) in 18.3%, and fluorescence in-situ hybridization (FISH) in 15.9% in the adenocarcinoma subgroup. For specimens tested with multiple methods, the consistency rates confirmed by IHC-VENTANA-D5F3 were 98.0% (822/839) for FISH, 98.7% (1,222/1,238) for NGS, and 91.3% (146/160) for RT-PCR. The overall ALK gene rearrangement rates were higher in females, patients of ≤ 35 years old, never smokers, tumor cellularity of > 50, and metastatic specimens used for testing in the total NSCLC population and adenocarcinoma subgroup (all P < 0.05).</p><p><strong>Conclusions: </strong>This study highlights the real-world variability and challenges of ALK test in advanced NSCLC, demonstrating a predominant use of IHC-VENTANA-D5F3 with high consistency and distinct clinicopathological features in ALK-positive patients. These findings underscore the need for a consensus on optimal test practices and support the development of refined ALK test strategies to enhance diagnostic accuracy and therapeutic decision-making in NSCLC.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141626091","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
Central nervous system efficacy of aumolertinib versus gefitinib in patients with untreated, EGFR-mutated, advanced non-small cell lung cancer: data from a randomized phase III trial (AENEAS). 未经治疗的表皮生长因子受体(EGFR)突变晚期非小细胞肺癌患者服用奥莫拉替尼和吉非替尼对中枢神经系统的疗效:一项随机III期试验(AENEAS)的数据。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-07-17 DOI: 10.1002/cac2.12594
Shun Lu, Xiaorong Dong, Hong Jian, Jianhua Chen, Gongyan Chen, Yuping Sun, Yinghua Ji, Ziping Wang, Jianhua Shi, Junguo Lu, Shaoshui Chen, Dongqing Lv, Guojun Zhang, Chunling Liu, Juan Li, Xinmin Yu, Zhong Lin, Zhuang Yu, Zhehai Wang, Jiuwei Cui, Xingxiang Xu, Jian Fang, Jifeng Feng, Zhi Xu, Rui Ma, Jie Hu, Nong Yang, Xiangdong Zhou, Xiaohong Wu, Chengping Hu, Zhihong Zhang, You Lu, Yanping Hu, Liyan Jiang, Qiming Wang, Renhua Guo, Jianying Zhou, Baolan Li, Chunhong Hu, Wancheng Tong, Helong Zhang, Lin Ma, Yuan Chen, Zhijun Jie, Yu Yao, Longzhen Zhang, Jie Weng, Weidong Li, Jianping Xiong, Xianwei Ye, Jianchun Duan, Haihua Yang, Meili Sun, Hongying Wei, Jiawei Wei, Zheyu Zhang, Qiong Wu

Background: The initial randomized, double-blinded, actively controlled, phase III ANEAS study (NCT03849768) demonstrated that aumolertinib showed superior efficacy relative to gefitinib as first-line therapy in epidermal growth factor receptor (EGFR)-mutated advanced non-small cell lung cancer (NSCLC). Metastatic disease in the central nervous system (CNS) remains a challenge in the management of NSCLC. This study aimed to compare the efficacy of aumolertinib versus gefitinib among patients with baseline CNS metastases in the ANEAS study.

Methods: Eligible patients were enrolled and randomly assigned in a 1:1 ratio to orally receive either aumolertinib or gefitinib in a double-blinded fashion. Patients with asymptomatic, stable CNS metastases were included. Follow-up imaging of the same modality as the initial CNS imaging was performed every 6 weeks for 15 months, then every 12 weeks. CNS response was assessed by a neuroradiological blinded, independent central review (neuroradiological-BICR). The primary endpoint for this subgroup analysis was CNS progression-free survival (PFS).

Results: Of the 429 patients enrolled and randomized in the ANEAS study, 106 patients were found to have CNS metastases (CNS Full Analysis Set, cFAS) at baseline by neuroradiological-BICR, and 60 of them had CNS target lesions (CNS Evaluable for Response, cEFR). Treatment with aumolertinib significantly prolonged median CNS PFS compared with gefitinib in both cFAS (29.0 vs. 8.3 months; hazard ratio [HR] = 0.31; 95% confidence interval [CI], 0.17-0.56; P < 0.001) and cEFR (29.0 vs. 8.3 months; HR = 0.26; 95% CI, 0.11-0.57; P < 0.001). The confirmed CNS overall response rate in cEFR was 85.7% and 75.0% in patients treated with aumolertinib and gefitinib, respectively. Competing risk analysis showed that the estimated probability of CNS progression without prior non-CNS progression or death was consistently lower with aumolertinib than with gefitinib in patients with and without CNS metastases at baseline. No new safety findings were observed.

Conclusions: These results indicate a potential advantage of aumolertinib over gefitinib in terms of CNS PFS and the risk of CNS progression in patients with EGFR-mutated advanced NSCLC with baseline CNS metastases.

Trial registration: ClinicalTrials.gov number, NCT03849768.

研究背景最初的随机、双盲、积极对照的III期ANEAS研究(NCT03849768)表明,奥美乐替尼作为表皮生长因子受体(EGFR)突变的晚期非小细胞肺癌(NSCLC)的一线疗法,疗效优于吉非替尼。中枢神经系统(CNS)转移性疾病仍然是治疗NSCLC的一大挑战。本研究旨在比较ANEAS研究中基线中枢神经系统转移患者中奥莫拉替尼和吉非替尼的疗效:符合条件的患者被纳入研究,并以1:1的比例随机分配到口服奥美替尼或吉非替尼的双盲研究中。研究对象包括无症状、病情稳定的中枢神经系统转移患者。在15个月内,每6周进行一次与首次中枢神经系统成像相同的随访成像,之后每12周进行一次。中枢神经系统反应由神经放射学盲法独立中央审查(神经放射学-BICR)评估。该亚组分析的主要终点是中枢神经系统无进展生存期(PFS):在ANEAS研究中随机登记的429名患者中,有106名患者在基线时被神经放射学-BICR发现有中枢神经系统转移(中枢神经系统全分析集,cFAS),其中60名患者有中枢神经系统靶病变(中枢神经系统可评估反应,cEFR)。在cFAS(29.0个月对8.3个月;危险比[HR] = 0.31;95%置信区间[CI],0.17-0.56;P <0.001)和cEFR(29.0个月对8.3个月;HR = 0.26;95% CI,0.11-0.57;P <0.001)中,与吉非替尼相比,奥莫勒替尼治疗显著延长了中位CNS PFS。在cEFR中,接受奥美替尼和吉非替尼治疗的患者中枢神经系统总应答率分别为85.7%和75.0%。竞争风险分析表明,在基线有中枢神经系统转移灶和无中枢神经系统转移灶的患者中,使用奥莫拉替尼而无既往非中枢神经系统进展或死亡的中枢神经系统进展的估计概率一直低于吉非替尼。没有观察到新的安全性结果:这些结果表明,对于基线有中枢神经系统转移的表皮生长因子受体(EGFR)突变晚期NSCLC患者,奥莫拉替尼在中枢神经系统PFS和中枢神经系统进展风险方面比吉非替尼具有潜在优势:试验注册:ClinicalTrials.gov 编号:NCT03849768。
{"title":"Central nervous system efficacy of aumolertinib versus gefitinib in patients with untreated, EGFR-mutated, advanced non-small cell lung cancer: data from a randomized phase III trial (AENEAS).","authors":"Shun Lu, Xiaorong Dong, Hong Jian, Jianhua Chen, Gongyan Chen, Yuping Sun, Yinghua Ji, Ziping Wang, Jianhua Shi, Junguo Lu, Shaoshui Chen, Dongqing Lv, Guojun Zhang, Chunling Liu, Juan Li, Xinmin Yu, Zhong Lin, Zhuang Yu, Zhehai Wang, Jiuwei Cui, Xingxiang Xu, Jian Fang, Jifeng Feng, Zhi Xu, Rui Ma, Jie Hu, Nong Yang, Xiangdong Zhou, Xiaohong Wu, Chengping Hu, Zhihong Zhang, You Lu, Yanping Hu, Liyan Jiang, Qiming Wang, Renhua Guo, Jianying Zhou, Baolan Li, Chunhong Hu, Wancheng Tong, Helong Zhang, Lin Ma, Yuan Chen, Zhijun Jie, Yu Yao, Longzhen Zhang, Jie Weng, Weidong Li, Jianping Xiong, Xianwei Ye, Jianchun Duan, Haihua Yang, Meili Sun, Hongying Wei, Jiawei Wei, Zheyu Zhang, Qiong Wu","doi":"10.1002/cac2.12594","DOIUrl":"https://doi.org/10.1002/cac2.12594","url":null,"abstract":"<p><strong>Background: </strong>The initial randomized, double-blinded, actively controlled, phase III ANEAS study (NCT03849768) demonstrated that aumolertinib showed superior efficacy relative to gefitinib as first-line therapy in epidermal growth factor receptor (EGFR)-mutated advanced non-small cell lung cancer (NSCLC). Metastatic disease in the central nervous system (CNS) remains a challenge in the management of NSCLC. This study aimed to compare the efficacy of aumolertinib versus gefitinib among patients with baseline CNS metastases in the ANEAS study.</p><p><strong>Methods: </strong>Eligible patients were enrolled and randomly assigned in a 1:1 ratio to orally receive either aumolertinib or gefitinib in a double-blinded fashion. Patients with asymptomatic, stable CNS metastases were included. Follow-up imaging of the same modality as the initial CNS imaging was performed every 6 weeks for 15 months, then every 12 weeks. CNS response was assessed by a neuroradiological blinded, independent central review (neuroradiological-BICR). The primary endpoint for this subgroup analysis was CNS progression-free survival (PFS).</p><p><strong>Results: </strong>Of the 429 patients enrolled and randomized in the ANEAS study, 106 patients were found to have CNS metastases (CNS Full Analysis Set, cFAS) at baseline by neuroradiological-BICR, and 60 of them had CNS target lesions (CNS Evaluable for Response, cEFR). Treatment with aumolertinib significantly prolonged median CNS PFS compared with gefitinib in both cFAS (29.0 vs. 8.3 months; hazard ratio [HR] = 0.31; 95% confidence interval [CI], 0.17-0.56; P < 0.001) and cEFR (29.0 vs. 8.3 months; HR = 0.26; 95% CI, 0.11-0.57; P < 0.001). The confirmed CNS overall response rate in cEFR was 85.7% and 75.0% in patients treated with aumolertinib and gefitinib, respectively. Competing risk analysis showed that the estimated probability of CNS progression without prior non-CNS progression or death was consistently lower with aumolertinib than with gefitinib in patients with and without CNS metastases at baseline. No new safety findings were observed.</p><p><strong>Conclusions: </strong>These results indicate a potential advantage of aumolertinib over gefitinib in terms of CNS PFS and the risk of CNS progression in patients with EGFR-mutated advanced NSCLC with baseline CNS metastases.</p><p><strong>Trial registration: </strong>ClinicalTrials.gov number, NCT03849768.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141626090","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
Immune mediated support of metastasis: Implication for bone invasion. 免疫介导的转移支持:对骨侵袭的影响
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-07-14 DOI: 10.1002/cac2.12584
Zengfeng Xin, Luying Qin, Yang Tang, Siyu Guo, Fangfang Li, Yuan Fang, Gege Li, Yihan Yao, Binbin Zheng, Bicheng Zhang, Dang Wu, Jie Xiao, Chao Ni, Qichun Wei, Ting Zhang

Bone is a common organ affected by metastasis in various advanced cancers, including lung, breast, prostate, colorectal, and melanoma. Once a patient is diagnosed with bone metastasis, the patient's quality of life and overall survival are significantly reduced owing to a wide range of morbidities and the increasing difficulty of treatment. Many studies have shown that bone metastasis is closely related to bone microenvironment, especially bone immune microenvironment. However, the effects of various immune cells in the bone microenvironment on bone metastasis remain unclear. Here, we described the changes in various immune cells during bone metastasis and discussed their related mechanisms. Osteoblasts, adipocytes, and other non-immune cells closely related to bone metastasis were also included. This review also summarized the existing treatment methods and potential therapeutic targets, and provided insights for future studies of cancer bone metastasis.

在各种晚期癌症(包括肺癌、乳腺癌、前列腺癌、结直肠癌和黑色素瘤)中,骨是受转移影响的常见器官。一旦确诊为骨转移,患者的生活质量和总体生存期都会因各种病症和治疗难度的增加而大大降低。许多研究表明,骨转移与骨微环境,尤其是骨免疫微环境密切相关。然而,骨微环境中各种免疫细胞对骨转移的影响仍不清楚。在此,我们描述了骨转移过程中各种免疫细胞的变化,并探讨了其相关机制。此外,还包括成骨细胞、脂肪细胞和其他与骨转移密切相关的非免疫细胞。本综述还总结了现有的治疗方法和潜在的治疗靶点,并为今后的癌症骨转移研究提供了启示。
{"title":"Immune mediated support of metastasis: Implication for bone invasion.","authors":"Zengfeng Xin, Luying Qin, Yang Tang, Siyu Guo, Fangfang Li, Yuan Fang, Gege Li, Yihan Yao, Binbin Zheng, Bicheng Zhang, Dang Wu, Jie Xiao, Chao Ni, Qichun Wei, Ting Zhang","doi":"10.1002/cac2.12584","DOIUrl":"https://doi.org/10.1002/cac2.12584","url":null,"abstract":"<p><p>Bone is a common organ affected by metastasis in various advanced cancers, including lung, breast, prostate, colorectal, and melanoma. Once a patient is diagnosed with bone metastasis, the patient's quality of life and overall survival are significantly reduced owing to a wide range of morbidities and the increasing difficulty of treatment. Many studies have shown that bone metastasis is closely related to bone microenvironment, especially bone immune microenvironment. However, the effects of various immune cells in the bone microenvironment on bone metastasis remain unclear. Here, we described the changes in various immune cells during bone metastasis and discussed their related mechanisms. Osteoblasts, adipocytes, and other non-immune cells closely related to bone metastasis were also included. This review also summarized the existing treatment methods and potential therapeutic targets, and provided insights for future studies of cancer bone metastasis.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-07-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141603313","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
Cellular senescence and metabolic reprogramming: Unraveling the intricate crosstalk in the immunosuppressive tumor microenvironment. 细胞衰老与代谢重编程:揭开免疫抑制性肿瘤微环境中错综复杂的相互关系。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-07-12 DOI: 10.1002/cac2.12591
Fusheng Zhang, Junchen Guo, Shengmiao Yu, Youwei Zheng, Meiqi Duan, Liang Zhao, Yihan Wang, Zhi Yang, Xiaofeng Jiang

The intrinsic oncogenic mechanisms and properties of the tumor microenvironment (TME) have been extensively investigated. Primary features of the TME include metabolic reprogramming, hypoxia, chronic inflammation, and tumor immunosuppression. Previous studies suggest that senescence-associated secretory phenotypes that mediate intercellular information exchange play a role in the dynamic evolution of the TME. Specifically, hypoxic adaptation, metabolic dysregulation, and phenotypic shifts in immune cells regulated by cellular senescence synergistically contribute to the development of an immunosuppressive microenvironment and chronic inflammation, thereby promoting the progression of tumor events. This review provides a comprehensive summary of the processes by which cellular senescence regulates the dynamic evolution of the tumor-adapted TME, with focus on the complex mechanisms underlying the relationship between senescence and changes in the biological functions of tumor cells. The available findings suggest that components of the TME collectively contribute to the progression of tumor events. The potential applications and challenges of targeted cellular senescence-based and combination therapies in clinical settings are further discussed within the context of advancing cellular senescence-related research.

人们对肿瘤微环境(TME)的内在致癌机制和特性进行了广泛研究。肿瘤微环境的主要特征包括代谢重编程、缺氧、慢性炎症和肿瘤免疫抑制。以往的研究表明,衰老相关的分泌表型介导了细胞间的信息交流,在 TME 的动态演化过程中发挥了作用。具体来说,缺氧适应、代谢失调以及由细胞衰老调控的免疫细胞表型转变协同促进了免疫抑制微环境和慢性炎症的发展,从而推动了肿瘤事件的进展。本综述全面总结了细胞衰老调控肿瘤适应性 TME 动态演化的过程,重点探讨了衰老与肿瘤细胞生物功能变化之间关系的复杂机制。现有研究结果表明,肿瘤适应性细胞生长环境的组成部分共同促成了肿瘤事件的进展。在推进细胞衰老相关研究的背景下,进一步讨论了基于细胞衰老的靶向疗法和综合疗法在临床环境中的潜在应用和挑战。
{"title":"Cellular senescence and metabolic reprogramming: Unraveling the intricate crosstalk in the immunosuppressive tumor microenvironment.","authors":"Fusheng Zhang, Junchen Guo, Shengmiao Yu, Youwei Zheng, Meiqi Duan, Liang Zhao, Yihan Wang, Zhi Yang, Xiaofeng Jiang","doi":"10.1002/cac2.12591","DOIUrl":"https://doi.org/10.1002/cac2.12591","url":null,"abstract":"<p><p>The intrinsic oncogenic mechanisms and properties of the tumor microenvironment (TME) have been extensively investigated. Primary features of the TME include metabolic reprogramming, hypoxia, chronic inflammation, and tumor immunosuppression. Previous studies suggest that senescence-associated secretory phenotypes that mediate intercellular information exchange play a role in the dynamic evolution of the TME. Specifically, hypoxic adaptation, metabolic dysregulation, and phenotypic shifts in immune cells regulated by cellular senescence synergistically contribute to the development of an immunosuppressive microenvironment and chronic inflammation, thereby promoting the progression of tumor events. This review provides a comprehensive summary of the processes by which cellular senescence regulates the dynamic evolution of the tumor-adapted TME, with focus on the complex mechanisms underlying the relationship between senescence and changes in the biological functions of tumor cells. The available findings suggest that components of the TME collectively contribute to the progression of tumor events. The potential applications and challenges of targeted cellular senescence-based and combination therapies in clinical settings are further discussed within the context of advancing cellular senescence-related research.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-07-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141598537","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
Defining and tracing subtypes of patient-derived xenograft models in pancreatic ductal adenocarcinoma 定义和追踪胰腺导管腺癌患者来源异种移植模型的亚型。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-07-10 DOI: 10.1002/cac2.12585
Sangyeop Hyun, Youngmin Han, Jae Yun Moon, Young-Ah Suh, Won-Gun Yun, Wooil Kwon, Jong-Eun Lee, Daeun Kim, Ja-Lok Ku, Jin-Young Jang, Daechan Park

Patient-derived xenograft (PDX) models have been used to explore therapeutic opportunities for pancreatic ductal adenocarcinoma (PDAC) [1]. Although original tumor characteristics are altered by cancer-stromal interactions in a PDX-specific manner [2], the implications of clonal evolution from PDAC tumors to PDX are largely unknown.

In this study, we have conducted a comprehensive genomic analysis using 36 patient-matched PDAC tumor and PDX samples (Figure 1A). The detailed methods regarding this study are described in the Supplementary Materials. The clinical information is summarized in Supplementary Table S1. To compare the somatic mutation profiles of PDAC tumors and PDX, 33 whole exome sequencing data were analyzed by using matched patient blood as a normal control. The proportion of PDX samples with Kirsten Rat Sarcoma Viral Oncogene Homolog (KRAS), Tumor Protein P53 (TP53), Mothers Against Decapentaplegic Homolog 4 (SMAD4), and cyclin-dependent kinase inhibitor 2A (CDKN2A) mutations increased compared to PDAC tumors, indicating that cancerous clones evolved in PDX from primary tumors (Supplementary Table S2-S5, Figure 1B) [3]. Specifically, the frequency of the KRAS G12D mutation increased during PDX establishment, suggesting that this mutation could be responsible for driving clonal evolution in PDX models (Supplementary Table S2). Next, we observed the high correlation of the variant allele frequencies (VAFs) of commonly mutated genes between matched PDAC tumors and PDX in pairwise comparison (Figure 1C), indicating that the overall mutation rate was conserved during PDX construction. When VAFs were compared at the gene level, VAFs of driver genes significantly increased in PDX compared to primary tumors (Figure 1D). Copy number variation (CNV) profiles of protein-coding genes were also similar between the matched samples (Figure 1E, Supplementary Figure S1), while the copy numbers of driver genes became more evident in PDX compared to primary tumors (Figure 1F). Clonality analysis showed that subclones of primary tumors evolved as monoclonal or polyclonal patterns in matched PDX (Supplementary Figure S2). Despite the lack of investigations into clonal evolution over passages, these results suggest that molecular subtypes of PDX could deviate from PDAC tumors via clonal evolution during PDX model construction.

To investigate whether conventional PDAC subtyping is applicable to PDX, the molecular subtypes defined by Bailey et al. [5] were assigned to PDAC tumors and PDX. PDAC tumors were clearly clustered according to the Bailey gene signatures, showing the worst prognosis of patients with the squamous subtype as previously reported (Figure 1G). However, PDX clustering based on the Bailey gene signatures exhibited 61% (22/36) conflicting subtypes between the matched PDAC tumor and PDX samples (Supplementary Table S6). In particular

患者衍生异种移植(PDX)模型已被用于探索胰腺导管腺癌(PDAC)的治疗机会[1]。在本研究中,我们利用 36 例患者匹配的 PDAC 肿瘤和 PDX 样本(图 1A)进行了全面的基因组分析。本研究的详细方法见补充材料。补充表 S1 总结了临床信息。为了比较 PDAC 肿瘤和 PDX 的体细胞突变情况,以匹配的患者血液作为正常对照,分析了 33 个全外显子组测序数据。与PDAC肿瘤相比,PDX样本中出现Kirsten鼠肉瘤病毒癌基因同源物(KRAS)、肿瘤蛋白P53(TP53)、母亲抗截瘫同源物4(SMAD4)和细胞周期蛋白依赖性激酶抑制剂2A(CDKN2A)突变的比例增加,表明PDX中的癌克隆是从原发肿瘤演化而来的(补充表S2-S5,图1B)[3]。具体而言,KRAS G12D 突变的频率在 PDX 建立过程中有所增加,表明该突变可能是 PDX 模型中克隆进化的驱动因素(补充表 S2)。接下来,我们观察到配对的 PDAC 肿瘤和 PDX 之间常见突变基因的变异等位基因频率(VAFs)在配对比较中的高度相关性(图 1C),表明在 PDX 构建过程中总体突变率是一致的。在基因水平上比较VAF时,与原发肿瘤相比,PDX中驱动基因的VAF显著增加(图1D)。配对样本间蛋白编码基因的拷贝数变异(CNV)图谱也相似(图 1E,补充图 S1),而 PDX 中驱动基因的拷贝数比原发肿瘤更明显(图 1F)。克隆性分析表明,原发性肿瘤的亚克隆在匹配的 PDX 中演变为单克隆或多克隆模式(补充图 S2)。为了研究传统的PDAC亚型是否适用于PDX,我们将Bailey等人[5]定义的分子亚型分配给了PDAC肿瘤和PDX。根据Bailey基因特征,PDAC肿瘤被清晰地聚类,如之前报道的那样,鳞状亚型患者的预后最差(图1G)。然而,基于贝利基因特征的 PDX 聚类显示,配对的 PDAC 肿瘤和 PDX 样本中有 61%(22/36)的亚型相互冲突(补充表 S6)。特别是,异常分化的内分泌外分泌亚型(ADEX)(n = 4)和免疫原性亚型(n = 2)的比例降低,这两种亚型的基因表达没有明显区别(图 1H),暗示了 PDX 中基质转换的影响。由于贝利亚型对PDX无效,我们定义了三种PDX特异性分子亚型(图1I,补充图S3,补充表S7)。基因本体分析表明,在贝利亚型中,簇1特征与细胞外基质组织有关,而簇2和簇3分别与鳞状细胞和胰腺原细胞相似(补充图S4)。特别是,PDX 第 2 群表现出与快速克隆扩增相关的基因表达特征的富集,如缺氧、糖酵解和 DNA 复制(补充图 S5)。虽然基于三种 PDX 亚型的生存率差异没有统计学意义,但群集 2 患者的预后表现出更明显的差异(图 1I),且术后 6 个月内早期复发(补充表 S8)。为了比较 PDX 亚型与人类亚型基因特征的相似性,我们从以往的研究[5, 4, 6]中收集了基质型、基底型和经典型等 3 种人系的 12 个不同特征,然后进行了基因重叠分析(图 1J)。热图中的数字代表了PDX亚型与人类亚型之间共享特征基因的数量,其中群组1、2和3分别对应基质、基底和经典系。根据这些结果,PDX 亚型的命名被归入三个群组:PDX-基质型、PDX-基底型和PDX-经典型。耐人寻味的是,ADEX亚型和免疫原性亚型与这三种PDX亚型没有任何重叠的基因特征。
{"title":"Defining and tracing subtypes of patient-derived xenograft models in pancreatic ductal adenocarcinoma","authors":"Sangyeop Hyun,&nbsp;Youngmin Han,&nbsp;Jae Yun Moon,&nbsp;Young-Ah Suh,&nbsp;Won-Gun Yun,&nbsp;Wooil Kwon,&nbsp;Jong-Eun Lee,&nbsp;Daeun Kim,&nbsp;Ja-Lok Ku,&nbsp;Jin-Young Jang,&nbsp;Daechan Park","doi":"10.1002/cac2.12585","DOIUrl":"10.1002/cac2.12585","url":null,"abstract":"<p>Patient-derived xenograft (PDX) models have been used to explore therapeutic opportunities for pancreatic ductal adenocarcinoma (PDAC) [<span>1</span>]. Although original tumor characteristics are altered by cancer-stromal interactions in a PDX-specific manner [<span>2</span>], the implications of clonal evolution from PDAC tumors to PDX are largely unknown.</p><p>In this study, we have conducted a comprehensive genomic analysis using 36 patient-matched PDAC tumor and PDX samples (Figure 1A). The detailed methods regarding this study are described in the Supplementary Materials. The clinical information is summarized in Supplementary Table S1. To compare the somatic mutation profiles of PDAC tumors and PDX, 33 whole exome sequencing data were analyzed by using matched patient blood as a normal control. The proportion of PDX samples with Kirsten Rat Sarcoma Viral Oncogene Homolog (<i>KRAS</i>), Tumor Protein P53 (<i>TP53</i>), Mothers Against Decapentaplegic Homolog 4 (<i>SMAD4</i>), and cyclin-dependent kinase inhibitor 2A (<i>CDKN2A</i>) mutations increased compared to PDAC tumors, indicating that cancerous clones evolved in PDX from primary tumors (Supplementary Table S2-S5, Figure 1B) [<span>3</span>]. Specifically, the frequency of the <i>KRAS</i> G12D mutation increased during PDX establishment, suggesting that this mutation could be responsible for driving clonal evolution in PDX models (Supplementary Table S2). Next, we observed the high correlation of the variant allele frequencies (VAFs) of commonly mutated genes between matched PDAC tumors and PDX in pairwise comparison (Figure 1C), indicating that the overall mutation rate was conserved during PDX construction. When VAFs were compared at the gene level, VAFs of driver genes significantly increased in PDX compared to primary tumors (Figure 1D). Copy number variation (CNV) profiles of protein-coding genes were also similar between the matched samples (Figure 1E, Supplementary Figure S1), while the copy numbers of driver genes became more evident in PDX compared to primary tumors (Figure 1F). Clonality analysis showed that subclones of primary tumors evolved as monoclonal or polyclonal patterns in matched PDX (Supplementary Figure S2). Despite the lack of investigations into clonal evolution over passages, these results suggest that molecular subtypes of PDX could deviate from PDAC tumors via clonal evolution during PDX model construction.</p><p>To investigate whether conventional PDAC subtyping is applicable to PDX, the molecular subtypes defined by Bailey <i>et al.</i> [<span>5</span>] were assigned to PDAC tumors and PDX. PDAC tumors were clearly clustered according to the Bailey gene signatures, showing the worst prognosis of patients with the squamous subtype as previously reported (Figure 1G). However, PDX clustering based on the Bailey gene signatures exhibited 61% (22/36) conflicting subtypes between the matched PDAC tumor and PDX samples (Supplementary Table S6). In particular","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12585","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141562645","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
Dual inhibition of sirtuins 1 and 2: reprogramming metabolic energy dynamics in chronic myeloid leukemia as an immunogenic anticancer strategy 双重抑制 sirtuins 1 和 2:重新规划慢性髓性白血病的代谢能量动态,作为一种免疫原性抗癌策略。
IF 20.1 1区 医学 Q1 ONCOLOGY Pub Date : 2024-07-08 DOI: 10.1002/cac2.12590
Michael Schnekenburger, Anne Lorant, Sruthi Reddy Gajulapalli, Ridhika Rajora, Jin-Young Lee, Aloran Mazumder, Haeun Yang, Christo Christov, Hyoung Jin Kang, Bernard Pirotte, Marc Diederich

Chronic myeloid leukemia (CML) is a lethal hematopoietic malignancy with a global incidence primarily attributed to the breakpoint cluster region-Abelson (BCR-ABL1) fusion oncogene in over 95% of cases. The introduction of tyrosine kinase inhibitors (TKIs) has revolutionized CML management; however, a subset of patients encounters challenges such as resistance and relapse, hindering the achievement of complete remission. Overcoming these challenges in CML also requires addressing persistent leukemia stem cells (LSCs) with inherent resistance mechanisms. Key regulators of LSC metabolism, proliferation, and survival, as well as genetic and epigenetic alterations, provide potential targets [1].

In this study, leveraging in silico analysis (methods and descriptions of other assays are in the Supplementary file) of LSCs from CML patients at diagnosis, we demonstrated enrichment in pathways predominantly associated with proliferation, oxidative phosphorylation (OXPHOS), and metabolism, concurrently with a decrease in immune response pathways (Figure 1A). Similarly, genes negatively impacting proliferation in CML cell lines, when depleted by CRISPR, were enriched in processes related to OXPHOS, metabolism, and proliferation, mirroring the enrichment observed in LSCs (Figure 1B) [2].

Sirtuins (SIRTs) are nicotinamide adenine dinucleotide (NAD)+-dependent histone deacetylases. SIRT1 and SIRT2 modulate key signaling proteins impacting metabolism, survival, and stress response [3]. Overexpression of SIRT1 and SIRT2 was observed in various cancers, including leukemia (Supplementary Figure S1A) [4, 5]. However, our analysis of CML patients revealed variability in the expression levels of SIRT1 and SIRT2 across datasets (Supplementary Figure S1B). Given the relatively small sample sizes, we recognized the limitations of relying solely on single gene expression data, as it may not fully capture the functional relevance of SIRT1/2 in CML. In response to this limitation, we expanded our analysis to identify broader gene expression patterns associated with SIRT1/2. Specifically, we identified a CML-related network comprising 180 co-regulated transcriptional targets associated with SIRT1/2 enriched in genes relevant to leukemia (Figure 1C, Supplementary Figure S1C-E). To quantify their collective impact, we consolidated the expression of all transcripts in the SIRT1/2 regulon into a unified score, referred to as the SIRT1-2 regulon score. This score effectively discriminated between healthy hematopoietic stem cells and LSCs from CML patients at diagnosis (Figure 1D), indicating the collective impact of SIRT1 and SIRT2 on the disease.

Given the complementary roles of SIRT1 and 2 in regulating metabolic and survival pathways and their potential to compensate for each other's loss of function [3], we postulated that exploiting metabolic vulnerabilities in LSCs throu

对差异表达的基因进行了分析,结果显示,参与增殖的基因下调,富集于受关键转录因子(如 MYC、JUN 和 E2F4)调控的基因,这些转录因子与 SIRT1/2 调节子有广泛联系;与免疫反应和细胞死亡机制相关的基因上调(图 1I,补充图 S12B-H、S13)。代谢反应富集分析显示,在 24 小时内,Si-711 会诱导 CML 细胞发生显著的代谢改变,包括糖酵解上调、核苷酸代谢和线粒体呼吸复合物下调(补充图 S12I-Q)。细胞内研究显示,SIRT1/2抑制诱导了CML中关键的代谢改变,导致耗氧率(OCR)降低、细胞外酸化率(ECAR)短暂增加,并最终导致整体代谢活性降低(图1J,补充图S14A-G)。作为对抑制 OCR 的适应性反应,Si-711 增强了糖酵解,导致细胞外酸化增加、乳酸释放以及 CML 细胞转向主要依赖糖酵解产生 ATP,偏离了它们最初对 OXPHOS 依赖性 ATP 的依赖(图 1K-M,补充图 S8B、S14H-K、S15A),这是 LSCs 和耐药细胞的特征[7]。最终,这些代谢改变导致总 ATP 含量逐渐耗竭,NAD+/NADH 比值发生显著变化,线粒体形态发生改变,包括洋葱状漩涡核心被肿胀的空间包围,同时嵴结构不那么凝结和无序(图 1N,补图 S8C、S14L、S15B-C、S16-S18)。此外,Si-711 处理增加了线粒体的超氧化物水平,表明诱导了氧化应激(图 1O,补充图 S19)。长期暴露后,在 5-10 µmol/L 的浓度范围内,所导致的能量崩溃会引导大多数髓系白血病模型的命运走向坏死样细胞死亡,选择性地靶向癌细胞,选择性因子从 10 到 105 不等(图 1P,补充图 S8D、S20-S22,补充表 S6)。这种非典型细胞死亡与传统的抗 CML 疗法不同,有望克服典型的细胞凋亡耐药机制[8]。抑制 SIRT1/2 可诱导不依赖于 caspase/RIPK3、但依赖于 RIPK1/MLKL/PARP1 的细胞坏死(图 1Q,补充图 S23-S26)。这种坏死导致内质网钙储存逐渐耗竭,导致钙在线粒体中积聚,与线粒体通透性转换孔的打开有关(补充图 S24、S27-S30)。用Si-711抑制SIRT1/2可诱导CML细胞的免疫调节性坏死,显著促进损伤相关分子模式(DAMPs)的释放,包括钙网蛋白、高迁移率基团框1(HMGB1)和ATP(图1R-U,补充图S31A-E)。这有可能引发免疫原性细胞死亡(ICD)并增强抗癌免疫反应[9]。值得注意的是,Si-711 处理促进了吞噬作用(图 1V,补充图 S31F),表明免疫反应可能会增强。我们进行了体内疫苗接种试验,这被认为是验证诱导 ICD 和评估治疗潜力的黄金标准 [10]。在肿瘤挑战下,接种了Si-711处理细胞的小鼠表现出肿瘤体积、肿瘤重量和脾肿大的显著减少(图1W,补充图S32-S33)。重要的是,Si-711诱导ICD的效果与已知的ICD诱导剂奥沙利铂相当。SIRT1和SIRT2的双重靶向可以通过代谢重编程和诱导免疫性坏死样细胞死亡来消灭LSCs。这种综合方法有望解决CML耐药和复发等难题,为改善患者预后提供新途径。Michael Schnekenburger进行了体外/细胞内实验;Sruthi Reddy Gajulapalli、Ridhika Rajora和Jin-Young Lee进行了体内实验;Hyoung Jin Kang提供了患者样本;Aloran Mazumder和Haeun Yang进行了额外的体外实验;Anne Lorant进行、分析和解释了生物信息数据和统计分析;Christo Christov解释和分析了组织学切片;Michael Schnekenburger和Marc Diederich构思和设计了该项目。Michael Schnekenburger、Anne Lorant和Marc Diederich撰写/编辑了手稿;Bernard Pirotte合成了抑制剂;Marc Diederich监督了该项目。所有作者均已阅读并批准手稿。
{"title":"Dual inhibition of sirtuins 1 and 2: reprogramming metabolic energy dynamics in chronic myeloid leukemia as an immunogenic anticancer strategy","authors":"Michael Schnekenburger,&nbsp;Anne Lorant,&nbsp;Sruthi Reddy Gajulapalli,&nbsp;Ridhika Rajora,&nbsp;Jin-Young Lee,&nbsp;Aloran Mazumder,&nbsp;Haeun Yang,&nbsp;Christo Christov,&nbsp;Hyoung Jin Kang,&nbsp;Bernard Pirotte,&nbsp;Marc Diederich","doi":"10.1002/cac2.12590","DOIUrl":"10.1002/cac2.12590","url":null,"abstract":"<p>Chronic myeloid leukemia (CML) is a lethal hematopoietic malignancy with a global incidence primarily attributed to the <i>breakpoint cluster region-Abelson</i> (BCR-ABL1) fusion oncogene in over 95% of cases. The introduction of tyrosine kinase inhibitors (TKIs) has revolutionized CML management; however, a subset of patients encounters challenges such as resistance and relapse, hindering the achievement of complete remission. Overcoming these challenges in CML also requires addressing persistent leukemia stem cells (LSCs) with inherent resistance mechanisms. Key regulators of LSC metabolism, proliferation, and survival, as well as genetic and epigenetic alterations, provide potential targets [<span>1</span>].</p><p>In this study, leveraging in silico analysis (methods and descriptions of other assays are in the Supplementary file) of LSCs from CML patients at diagnosis, we demonstrated enrichment in pathways predominantly associated with proliferation, oxidative phosphorylation (OXPHOS), and metabolism, concurrently with a decrease in immune response pathways (Figure 1A). Similarly, genes negatively impacting proliferation in CML cell lines, when depleted by CRISPR, were enriched in processes related to OXPHOS, metabolism, and proliferation, mirroring the enrichment observed in LSCs (Figure 1B) [<span>2</span>].</p><p>Sirtuins (SIRTs) are nicotinamide adenine dinucleotide (NAD)<sup>+</sup>-dependent histone deacetylases. SIRT1 and SIRT2 modulate key signaling proteins impacting metabolism, survival, and stress response [<span>3</span>]. Overexpression of SIRT1 and SIRT2 was observed in various cancers, including leukemia (Supplementary Figure S1A) [<span>4, 5</span>]. However, our analysis of CML patients revealed variability in the expression levels of SIRT1 and SIRT2 across datasets (Supplementary Figure S1B). Given the relatively small sample sizes, we recognized the limitations of relying solely on single gene expression data, as it may not fully capture the functional relevance of SIRT1/2 in CML. In response to this limitation, we expanded our analysis to identify broader gene expression patterns associated with SIRT1/2. Specifically, we identified a CML-related network comprising 180 co-regulated transcriptional targets associated with SIRT1/2 enriched in genes relevant to leukemia (Figure 1C, Supplementary Figure S1C-E). To quantify their collective impact, we consolidated the expression of all transcripts in the SIRT1/2 regulon into a unified score, referred to as the SIRT1-2 regulon score. This score effectively discriminated between healthy hematopoietic stem cells and LSCs from CML patients at diagnosis (Figure 1D), indicating the collective impact of SIRT1 and SIRT2 on the disease.</p><p>Given the complementary roles of SIRT1 and 2 in regulating metabolic and survival pathways and their potential to compensate for each other's loss of function [<span>3</span>], we postulated that exploiting metabolic vulnerabilities in LSCs throu","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":null,"pages":null},"PeriodicalIF":20.1,"publicationDate":"2024-07-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12590","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141554217","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
Cancer Communications
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