首页 > 最新文献

Cancer Communications最新文献

英文 中文
Targeting autophagy overcomes cancer-intrinsic resistance to CAR-T immunotherapy in B-cell malignancies 以自噬为靶点克服B细胞恶性肿瘤对CAR-T免疫疗法的内在抗药性。
IF 16.2 1区 医学 Q1 ONCOLOGY Pub Date : 2024-02-26 DOI: 10.1002/cac2.12525
Lu Tang, Huan Zhang, Fen Zhou, Qiuzhe Wei, Mengyi Du, Jianghua Wu, Chenggong Li, Wenjing Luo, Jie Zhou, Xindi Wang, Zhaozhao Chen, Yinqiang Zhang, Zhongpei Huang, Zhuolin Wu, Yuxi Wen, Huiwen Jiang, Danying Liao, Haiming Kou, Wei Xiong, Heng Mei, Yu Hu
<div> <section> <h3> Background</h3> <p>Chimeric antigen receptor T (CAR-T) therapy has substantially revolutionized the clinical outcomes of patients with hematologic malignancies, but the cancer-intrinsic mechanisms underlying resistance to CAR-T cells remain yet to be fully understood. This study aims to explore the molecular determinants of cancer cell sensitivity to CAR-T cell-mediated killing and to provide a better understanding of the underlying mechanisms and potential modulation to improve clinical efficacy.</p> </section> <section> <h3> Methods</h3> <p>The human whole-genome CRISPR/Cas9-based knockout screening was conducted to identify key genes that enable cancer cells to evade CD19 CAR-T-cell-mediated killing. The in vitro cytotoxicity assays and evaluation of tumor tissue and bone marrow specimens were further conducted to confirm the role of the key genes in cancer cell susceptibility to CAR-T cells. In addition, the specific mechanisms influencing CAR-T cell-mediated cancer clearance were elucidated in mouse and cellular models.</p> </section> <section> <h3> Results</h3> <p>The CRISPR/Cas9-based knockout screening showed that the enrichment of autophagy-related genes (<i>ATG3</i>, <i>BECN1</i>, and <i>RB1CC1</i>) provided protection of cancer cells from CD19 CAR-T cell-mediated cytotoxicity. These findings were further validated by in vitro cytotoxicity assays in cells with genetic and pharmacological inhibition of autophagy. Notably, higher expression of the three autophagy-related proteins in tumor samples was correlated with poorer responsiveness and worse survival in patients with relapsed/refractory B-cell lymphoma after CD19 CAR-T therapy. Bulk RNA sequencing analysis of bone marrow samples from B-cell leukemia patients also suggested the clinical relevance of autophagy to the therapeutic response and relapse after CD19 CAR-T cell therapy. Pharmacological inhibition of autophagy and knockout of RB1CC1 could dramatically sensitize tumor cells to CD19 CAR-T cell-mediated killing in mouse models of both B-cell leukemia and lymphoma. Moreover, our study revealed that cancer-intrinsic autophagy mediates evasion of CAR-T cells via the TNF-α-TNFR1 axis-mediated apoptosis and STAT1/IRF1-induced chemokine signaling activation.</p> </section> <section> <h3> Conclusions</h3> <p>These findings confirm that autophagy signaling in B-cell malignancies is essential for the effective cytotoxic function of CAR-T cells and thereby pave the way for the development of autophagy-targeting strategies to improve the clinical efficacy of CAR-T cell i
背景:嵌合抗原受体T(CAR-T)疗法极大地改变了血液恶性肿瘤患者的临床疗效,但CAR-T细胞耐药性的癌症内在机制仍有待全面了解。本研究旨在探索癌细胞对CAR-T细胞介导的杀伤敏感性的分子决定因素,从而更好地了解其潜在机制和可能的调控,以提高临床疗效:方法:通过基于CRISPR/Cas9的人类全基因组基因敲除筛选,找出使癌细胞逃避CD19 CAR-T细胞介导的杀伤的关键基因。通过体外细胞毒性实验以及对肿瘤组织和骨髓标本的评估,进一步证实了这些关键基因在癌细胞对 CAR-T 细胞易感性中的作用。此外,还在小鼠和细胞模型中阐明了影响CAR-T细胞介导的癌症清除的具体机制:结果:基于CRISPR/Cas9的基因敲除筛选显示,自噬相关基因(ATG3、BECN1和RB1CC1)的富集可保护癌细胞不受CD19 CAR-T细胞介导的细胞毒性的影响。对自噬进行基因和药物抑制的细胞进行体外细胞毒性实验进一步验证了这些发现。值得注意的是,三种自噬相关蛋白在肿瘤样本中的高表达与CD19 CAR-T疗法后复发/难治性B细胞淋巴瘤患者的较差反应性和较差存活率相关。对B细胞白血病患者骨髓样本的大量RNA测序分析也表明,自噬与CD19 CAR-T细胞疗法后的治疗反应和复发具有临床相关性。在B细胞白血病和淋巴瘤的小鼠模型中,药理抑制自噬和敲除RB1CC1可使肿瘤细胞对CD19 CAR-T细胞介导的杀伤显著敏感。此外,我们的研究还发现,癌症内在自噬通过 TNF-α-TNFR1 轴介导的细胞凋亡和 STAT1/IRF1 诱导的趋化因子信号激活,介导 CAR-T 细胞的逃避:这些发现证实了自噬信号在 B 细胞恶性肿瘤中对 CAR-T 细胞的有效细胞毒性功能至关重要,从而为开发自噬靶向策略以提高 CAR-T 细胞免疫疗法的临床疗效铺平了道路。
{"title":"Targeting autophagy overcomes cancer-intrinsic resistance to CAR-T immunotherapy in B-cell malignancies","authors":"Lu Tang,&nbsp;Huan Zhang,&nbsp;Fen Zhou,&nbsp;Qiuzhe Wei,&nbsp;Mengyi Du,&nbsp;Jianghua Wu,&nbsp;Chenggong Li,&nbsp;Wenjing Luo,&nbsp;Jie Zhou,&nbsp;Xindi Wang,&nbsp;Zhaozhao Chen,&nbsp;Yinqiang Zhang,&nbsp;Zhongpei Huang,&nbsp;Zhuolin Wu,&nbsp;Yuxi Wen,&nbsp;Huiwen Jiang,&nbsp;Danying Liao,&nbsp;Haiming Kou,&nbsp;Wei Xiong,&nbsp;Heng Mei,&nbsp;Yu Hu","doi":"10.1002/cac2.12525","DOIUrl":"10.1002/cac2.12525","url":null,"abstract":"&lt;div&gt;\u0000 \u0000 \u0000 &lt;section&gt;\u0000 \u0000 &lt;h3&gt; Background&lt;/h3&gt;\u0000 \u0000 &lt;p&gt;Chimeric antigen receptor T (CAR-T) therapy has substantially revolutionized the clinical outcomes of patients with hematologic malignancies, but the cancer-intrinsic mechanisms underlying resistance to CAR-T cells remain yet to be fully understood. This study aims to explore the molecular determinants of cancer cell sensitivity to CAR-T cell-mediated killing and to provide a better understanding of the underlying mechanisms and potential modulation to improve clinical efficacy.&lt;/p&gt;\u0000 &lt;/section&gt;\u0000 \u0000 &lt;section&gt;\u0000 \u0000 &lt;h3&gt; Methods&lt;/h3&gt;\u0000 \u0000 &lt;p&gt;The human whole-genome CRISPR/Cas9-based knockout screening was conducted to identify key genes that enable cancer cells to evade CD19 CAR-T-cell-mediated killing. The in vitro cytotoxicity assays and evaluation of tumor tissue and bone marrow specimens were further conducted to confirm the role of the key genes in cancer cell susceptibility to CAR-T cells. In addition, the specific mechanisms influencing CAR-T cell-mediated cancer clearance were elucidated in mouse and cellular models.&lt;/p&gt;\u0000 &lt;/section&gt;\u0000 \u0000 &lt;section&gt;\u0000 \u0000 &lt;h3&gt; Results&lt;/h3&gt;\u0000 \u0000 &lt;p&gt;The CRISPR/Cas9-based knockout screening showed that the enrichment of autophagy-related genes (&lt;i&gt;ATG3&lt;/i&gt;, &lt;i&gt;BECN1&lt;/i&gt;, and &lt;i&gt;RB1CC1&lt;/i&gt;) provided protection of cancer cells from CD19 CAR-T cell-mediated cytotoxicity. These findings were further validated by in vitro cytotoxicity assays in cells with genetic and pharmacological inhibition of autophagy. Notably, higher expression of the three autophagy-related proteins in tumor samples was correlated with poorer responsiveness and worse survival in patients with relapsed/refractory B-cell lymphoma after CD19 CAR-T therapy. Bulk RNA sequencing analysis of bone marrow samples from B-cell leukemia patients also suggested the clinical relevance of autophagy to the therapeutic response and relapse after CD19 CAR-T cell therapy. Pharmacological inhibition of autophagy and knockout of RB1CC1 could dramatically sensitize tumor cells to CD19 CAR-T cell-mediated killing in mouse models of both B-cell leukemia and lymphoma. Moreover, our study revealed that cancer-intrinsic autophagy mediates evasion of CAR-T cells via the TNF-α-TNFR1 axis-mediated apoptosis and STAT1/IRF1-induced chemokine signaling activation.&lt;/p&gt;\u0000 &lt;/section&gt;\u0000 \u0000 &lt;section&gt;\u0000 \u0000 &lt;h3&gt; Conclusions&lt;/h3&gt;\u0000 \u0000 &lt;p&gt;These findings confirm that autophagy signaling in B-cell malignancies is essential for the effective cytotoxic function of CAR-T cells and thereby pave the way for the development of autophagy-targeting strategies to improve the clinical efficacy of CAR-T cell i","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":"44 3","pages":"408-432"},"PeriodicalIF":16.2,"publicationDate":"2024-02-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12525","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139970976","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
SERPINE2 promotes liver cancer metastasis by inhibiting c-Cbl-mediated EGFR ubiquitination and degradation SERPINE2 通过抑制 c-Cbl 介导的表皮生长因子受体泛素化和降解,促进肝癌转移。
IF 16.2 1区 医学 Q1 ONCOLOGY Pub Date : 2024-02-26 DOI: 10.1002/cac2.12527
Shiyu Zhang, Xing Jia, Haojiang Dai, Xingxin Zhu, Wenfeng Song, Suchen Bian, Hao Wu, Shinuo Chen, Yangbo Tang, Junran Chen, Cheng Jin, Mengqiao Zhou, Haiyang Xie, Shusen Zheng, Penghong Song
<div> <section> <h3> Background</h3> <p>Liver cancer is a malignancy with high morbidity and mortality rates. Serpin family E member 2 (SERPINE2) has been reported to play a key role in the metastasis of many tumors. In this study, we aimed to investigate the potential mechanism of SERPINE2 in liver cancer metastasis.</p> </section> <section> <h3> Methods</h3> <p>The Cancer Genome Atlas database (TCGA), including DNA methylation and transcriptome sequencing data, was utilized to identify the crucial oncogene associated with DNA methylation and cancer progression in liver cancer. Data from the TCGA and RNA sequencing for 94 pairs of liver cancer tissues were used to explore the correlation between SERPINE2 expression and clinical parameters of patients. DNA methylation sequencing was used to detect the DNA methylation levels in liver cancer tissues and cells. RNA sequencing, cytokine assays, immunoprecipitation (IP) and mass spectrometry (MS) assays, protein stability assays, and ubiquitination assays were performed to explore the regulatory mechanism of SERPINE2 in liver cancer metastasis. Patient-derived xenografts and tumor organoid models were established to determine the role of SERPINE2 in the treatment of liver cancer using sorafenib.</p> </section> <section> <h3> Results</h3> <p>Based on the public database screening, SERPINE2 was identified as a tumor promoter regulated by DNA methylation. SERPINE2 expression was significantly higher in liver cancer tissues and was associated with the dismal prognosis in patients with liver cancer. SERPINE2 promoted liver cancer metastasis by enhancing cell pseudopodia formation, cell adhesion, cancer-associated fibroblast activation, extracellular matrix remodeling, and angiogenesis. IP/MS assays confirmed that SERPINE2 activated epidermal growth factor receptor (EGFR) and its downstream signaling pathways by interacting with EGFR. Mechanistically, SERPINE2 inhibited EGFR ubiquitination and maintained its protein stability by competing with the E3 ubiquitin ligase, c-Cbl. Additionally, EGFR was activated in liver cancer cells after sorafenib treatment, and SERPINE2 knockdown-induced EGFR downregulation significantly enhanced the therapeutic efficacy of sorafenib against liver cancer. Furthermore, we found that SERPINE2 knockdown also had a sensitizing effect on lenvatinib treatment.</p> </section> <section> <h3> Conclusions</h3> <p>SERPINE2 promoted liver cancer metastasis by preventing EGFR degradation via c-Cbl-mediated ubiquitination, suggesting that inhibition of the SERPINE2-EGFR axis may be a potenti
背景:肝癌是一种发病率和死亡率都很高的恶性肿瘤。据报道,丝氨酸蛋白家族 E 成员 2(SERPINE2)在许多肿瘤的转移中起着关键作用。本研究旨在探讨SERPINE2在肝癌转移中的潜在机制:方法:利用癌症基因组图谱数据库(TCGA),包括DNA甲基化和转录组测序数据,确定肝癌中与DNA甲基化和癌症进展相关的关键癌基因。研究人员利用 TCGA 数据和 94 对肝癌组织的 RNA 测序数据,探讨了 SERPINE2 表达与患者临床参数之间的相关性。DNA甲基化测序用于检测肝癌组织和细胞中的DNA甲基化水平。通过RNA测序、细胞因子检测、免疫沉淀(IP)和质谱(MS)检测、蛋白质稳定性检测和泛素化检测来探讨SERPINE2在肝癌转移中的调控机制。建立了患者来源的异种移植和肿瘤类器官模型,以确定SERPINE2在索拉非尼治疗肝癌中的作用:根据公共数据库筛选,SERPINE2被确定为受DNA甲基化调控的肿瘤启动子。SERPINE2在肝癌组织中的表达明显升高,并与肝癌患者的预后不良有关。SERPINE2通过增强细胞伪足形成、细胞粘附、癌相关成纤维细胞活化、细胞外基质重塑和血管生成,促进肝癌转移。IP/MS测定证实,SERPINE2通过与表皮生长因子受体(EGFR)相互作用,激活了表皮生长因子受体(EGFR)及其下游信号通路。从机理上讲,SERPINE2 通过与 E3 泛素连接酶 c-Cbl 竞争,抑制了表皮生长因子受体的泛素化,并保持了其蛋白质的稳定性。此外,索拉非尼治疗后肝癌细胞中的表皮生长因子受体被激活,而 SERPINE2 敲除诱导的表皮生长因子受体下调能显著增强索拉非尼对肝癌的疗效。此外,我们还发现敲除SERPINE2对来伐替尼治疗也有增敏作用:结论:SERPINE2通过c-Cbl介导的泛素化阻止表皮生长因子受体(EGFR)降解,从而促进肝癌转移。
{"title":"SERPINE2 promotes liver cancer metastasis by inhibiting c-Cbl-mediated EGFR ubiquitination and degradation","authors":"Shiyu Zhang,&nbsp;Xing Jia,&nbsp;Haojiang Dai,&nbsp;Xingxin Zhu,&nbsp;Wenfeng Song,&nbsp;Suchen Bian,&nbsp;Hao Wu,&nbsp;Shinuo Chen,&nbsp;Yangbo Tang,&nbsp;Junran Chen,&nbsp;Cheng Jin,&nbsp;Mengqiao Zhou,&nbsp;Haiyang Xie,&nbsp;Shusen Zheng,&nbsp;Penghong Song","doi":"10.1002/cac2.12527","DOIUrl":"10.1002/cac2.12527","url":null,"abstract":"&lt;div&gt;\u0000 \u0000 \u0000 &lt;section&gt;\u0000 \u0000 &lt;h3&gt; Background&lt;/h3&gt;\u0000 \u0000 &lt;p&gt;Liver cancer is a malignancy with high morbidity and mortality rates. Serpin family E member 2 (SERPINE2) has been reported to play a key role in the metastasis of many tumors. In this study, we aimed to investigate the potential mechanism of SERPINE2 in liver cancer metastasis.&lt;/p&gt;\u0000 &lt;/section&gt;\u0000 \u0000 &lt;section&gt;\u0000 \u0000 &lt;h3&gt; Methods&lt;/h3&gt;\u0000 \u0000 &lt;p&gt;The Cancer Genome Atlas database (TCGA), including DNA methylation and transcriptome sequencing data, was utilized to identify the crucial oncogene associated with DNA methylation and cancer progression in liver cancer. Data from the TCGA and RNA sequencing for 94 pairs of liver cancer tissues were used to explore the correlation between SERPINE2 expression and clinical parameters of patients. DNA methylation sequencing was used to detect the DNA methylation levels in liver cancer tissues and cells. RNA sequencing, cytokine assays, immunoprecipitation (IP) and mass spectrometry (MS) assays, protein stability assays, and ubiquitination assays were performed to explore the regulatory mechanism of SERPINE2 in liver cancer metastasis. Patient-derived xenografts and tumor organoid models were established to determine the role of SERPINE2 in the treatment of liver cancer using sorafenib.&lt;/p&gt;\u0000 &lt;/section&gt;\u0000 \u0000 &lt;section&gt;\u0000 \u0000 &lt;h3&gt; Results&lt;/h3&gt;\u0000 \u0000 &lt;p&gt;Based on the public database screening, SERPINE2 was identified as a tumor promoter regulated by DNA methylation. SERPINE2 expression was significantly higher in liver cancer tissues and was associated with the dismal prognosis in patients with liver cancer. SERPINE2 promoted liver cancer metastasis by enhancing cell pseudopodia formation, cell adhesion, cancer-associated fibroblast activation, extracellular matrix remodeling, and angiogenesis. IP/MS assays confirmed that SERPINE2 activated epidermal growth factor receptor (EGFR) and its downstream signaling pathways by interacting with EGFR. Mechanistically, SERPINE2 inhibited EGFR ubiquitination and maintained its protein stability by competing with the E3 ubiquitin ligase, c-Cbl. Additionally, EGFR was activated in liver cancer cells after sorafenib treatment, and SERPINE2 knockdown-induced EGFR downregulation significantly enhanced the therapeutic efficacy of sorafenib against liver cancer. Furthermore, we found that SERPINE2 knockdown also had a sensitizing effect on lenvatinib treatment.&lt;/p&gt;\u0000 &lt;/section&gt;\u0000 \u0000 &lt;section&gt;\u0000 \u0000 &lt;h3&gt; Conclusions&lt;/h3&gt;\u0000 \u0000 &lt;p&gt;SERPINE2 promoted liver cancer metastasis by preventing EGFR degradation via c-Cbl-mediated ubiquitination, suggesting that inhibition of the SERPINE2-EGFR axis may be a potenti","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":"44 3","pages":"384-407"},"PeriodicalIF":16.2,"publicationDate":"2024-02-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12527","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139970975","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
NAT10-mediated ac4C-modified ANKZF1 promotes tumor progression and lymphangiogenesis in clear-cell renal cell carcinoma by attenuating YWHAE-driven cytoplasmic retention of YAP1 NAT10 介导的 ac4 C 修饰的 ANKZF1 通过减弱 YWHAE 驱动的 YAP1 胞质滞留,促进透明细胞肾细胞癌的肿瘤进展和淋巴管生成。
IF 16.2 1区 医学 Q1 ONCOLOGY Pub Date : 2024-02-26 DOI: 10.1002/cac2.12523
Daojia Miao, Jian Shi, Qingyang Lv, Diaoyi Tan, Chuanyi Zhao, Zhiyong Xiong, Xiaoping Zhang

Background

Lymphatic metastasis is one of the most common metastatic routes and indicates a poor prognosis in clear-cell renal cell carcinoma (ccRCC). N-acetyltransferase 10 (NAT10) is known to catalyze N4-acetylcytidine (ac4C) modification of mRNA and participate in many cellular processes. However, its role in the lymphangiogenic process of ccRCC has not been reported. This study aimed to elucidate the role of NAT10 in ccRCC lymphangiogenesis, providing valuable insights into potential therapeutic targets for intervention.

Methods

ac4C modification and NAT10 expression levels in ccRCC were assessed using public databases and clinical samples. Functional investigations involved manipulating NAT10 expression in cellular and mouse models to study its role in ccRCC. Mechanistic insights were gained through a combination of RNA sequencing, mass spectrometry, co-immunoprecipitation, RNA immunoprecipitation, immunofluorescence, and site-specific mutation analyses.

Results

We found that ac4C modification and NAT10 expression levels increased in ccRCC. NAT10 promoted tumor progression and lymphangiogenesis of ccRCC by enhancing the nuclear import of Yes1-associated transcriptional regulator (YAP1). Subsequently, we identified ankyrin repeat and zinc finger peptidyl tRNA hydrolase 1 (ANKZF1) as the functional target of NAT10, and its upregulation in ccRCC was caused by NAT10-mediated ac4C modification. Mechanistic analyses demonstrated that ANKZF1 interacted with tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein epsilon (YWHAE) to competitively inhibit cytoplasmic retention of YAP1, leading to transcriptional activation of pro-lymphangiogenic factors.

Conclusions

These results suggested a pro-cancer role of NAT10-mediated acetylation in ccRCC and identified the NAT10/ANKZF1/YAP1 axis as an under-reported pathway involving tumor progression and lymphangiogenesis in ccRCC.

背景:淋巴转移是最常见的转移途径之一,预示着透明细胞肾细胞癌(ccRCC)的不良预后。众所周知,N-乙酰转移酶 10(NAT10)可催化 mRNA 的 N4-乙酰胞苷(ac4 C)修饰,并参与许多细胞过程。然而,它在ccRCC淋巴管生成过程中的作用尚未见报道。本研究旨在阐明NAT10在ccRCC淋巴管生成过程中的作用,为潜在的治疗靶点提供有价值的见解。方法:利用公共数据库和临床样本评估ccRCC中ac4 C修饰和NAT10的表达水平。功能研究包括在细胞和小鼠模型中操纵 NAT10 的表达,以研究其在 ccRCC 中的作用。通过结合 RNA 测序、质谱分析、共免疫沉淀、RNA 免疫沉淀、免疫荧光和位点特异性突变分析,我们获得了对机理的深入了解:结果:我们发现ac4 C修饰和NAT10在ccRCC中的表达水平升高。NAT10通过增强Yes1相关转录调节因子(YAP1)的核导入促进了ccRCC的肿瘤进展和淋巴管生成。随后,我们发现杏仁蛋白重复和锌指肽基tRNA水解酶1(ANKZF1)是NAT10的功能靶标,其在ccRCC中的上调是由NAT10介导的ac4 C修饰引起的。机理分析表明,ANKZF1与酪氨酸3-单氧化酶/色氨酸5-单氧化酶活化蛋白epsilon(YWHAE)相互作用,竞争性抑制YAP1的细胞质滞留,导致促淋巴管生成因子的转录激活:这些结果表明了NAT10介导的乙酰化在ccRCC中的促癌作用,并确定了NAT10/ANKZF1/YAP1轴是ccRCC中涉及肿瘤进展和淋巴管生成的一个未被充分报道的通路。
{"title":"NAT10-mediated ac4C-modified ANKZF1 promotes tumor progression and lymphangiogenesis in clear-cell renal cell carcinoma by attenuating YWHAE-driven cytoplasmic retention of YAP1","authors":"Daojia Miao,&nbsp;Jian Shi,&nbsp;Qingyang Lv,&nbsp;Diaoyi Tan,&nbsp;Chuanyi Zhao,&nbsp;Zhiyong Xiong,&nbsp;Xiaoping Zhang","doi":"10.1002/cac2.12523","DOIUrl":"10.1002/cac2.12523","url":null,"abstract":"<div>\u0000 \u0000 \u0000 <section>\u0000 \u0000 <h3> Background</h3>\u0000 \u0000 <p>Lymphatic metastasis is one of the most common metastatic routes and indicates a poor prognosis in clear-cell renal cell carcinoma (ccRCC). N-acetyltransferase 10 (NAT10) is known to catalyze N4-acetylcytidine (ac<sup>4</sup>C) modification of mRNA and participate in many cellular processes. However, its role in the lymphangiogenic process of ccRCC has not been reported. This study aimed to elucidate the role of NAT10 in ccRCC lymphangiogenesis, providing valuable insights into potential therapeutic targets for intervention.</p>\u0000 </section>\u0000 \u0000 <section>\u0000 \u0000 <h3> Methods</h3>\u0000 \u0000 <p>ac<sup>4</sup>C modification and NAT10 expression levels in ccRCC were assessed using public databases and clinical samples. Functional investigations involved manipulating NAT10 expression in cellular and mouse models to study its role in ccRCC. Mechanistic insights were gained through a combination of RNA sequencing, mass spectrometry, co-immunoprecipitation, RNA immunoprecipitation, immunofluorescence, and site-specific mutation analyses.</p>\u0000 </section>\u0000 \u0000 <section>\u0000 \u0000 <h3> Results</h3>\u0000 \u0000 <p>We found that ac<sup>4</sup>C modification and NAT10 expression levels increased in ccRCC. NAT10 promoted tumor progression and lymphangiogenesis of ccRCC by enhancing the nuclear import of Yes1-associated transcriptional regulator (YAP1). Subsequently, we identified ankyrin repeat and zinc finger peptidyl tRNA hydrolase 1 (ANKZF1) as the functional target of NAT10, and its upregulation in ccRCC was caused by NAT10-mediated ac<sup>4</sup>C modification. Mechanistic analyses demonstrated that ANKZF1 interacted with tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein epsilon (YWHAE) to competitively inhibit cytoplasmic retention of YAP1, leading to transcriptional activation of pro-lymphangiogenic factors.</p>\u0000 </section>\u0000 \u0000 <section>\u0000 \u0000 <h3> Conclusions</h3>\u0000 \u0000 <p>These results suggested a pro-cancer role of NAT10-mediated acetylation in ccRCC and identified the NAT10/ANKZF1/YAP1 axis as an under-reported pathway involving tumor progression and lymphangiogenesis in ccRCC.</p>\u0000 </section>\u0000 </div>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":"44 3","pages":"361-383"},"PeriodicalIF":16.2,"publicationDate":"2024-02-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12523","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139970974","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
A novel strategy for treating oncogene-mutated tumors by targeting tumor microenvironment and synergistically enhancing anti-PD-1 immunotherapy 通过靶向肿瘤微环境和协同增强抗 PD-1 免疫疗法治疗癌基因突变肿瘤的新策略。
IF 16.2 1区 医学 Q1 ONCOLOGY Pub Date : 2024-02-09 DOI: 10.1002/cac2.12521
Yingqiang Liu, Linjiang Tong, Mengge Zhang, Qi Zhang, Qiupei Liu, Fang Feng, Yan Li, Mengzhen Lai, Haotian Tang, Yi Chen, Meiyu Geng, Wenhu Duan, Jian Ding, Hua Xie
<p>Oncogenes are critical factors in tumorigenesis of diverse cancer types and play essential roles in tumor immune escape. Mutations in Kirsten rat sarcoma viral oncogene homolog (<i>KRAS</i>) and epidermal growth factor receptor (<i>EGFR</i>) are among the most frequent gain-of-function alterations [<span>1</span>]. After many years of in-depth research, inhibitors targeting <i>EGFR</i> or <i>KRAS</i> mutations have been successfully developed, however, their clinical benefit is relatively limited, and they will inevitably encounter the challenge of drug resistance. The emergence of resistance is attributed to secondary mutations in driver genes and other complicated factors. It is worth noting that approved treatment strategies are currently lacking for tumors with different types of <i>KRAS</i> or <i>EGFR</i> mutations, including <i>KRAS<sup>G12D</sup></i>, <i>KRAS<sup>G13D</sup></i>, and <i>EGFR<sup>C797S</sup></i> mutations that are common in tumors [<span>2</span>]. Additionally, oncogene mutations could trigger a cascade of tumor microenvironment changes, ultimately resulting in tumor progression or resistance to programmed death-1 (PD-1) antibody therapy [<span>3, 4</span>]. SYHA1813, a novel vascular endothelial growth factor receptor (VEGFR) and colony-stimulating factor 1 receptor (CSF1R) dual inhibitor, exhibited potent preclinical anti-glioma activity by inhibiting angiogenesis and promoting tumor immunity and showed promising efficacy in an ongoing clinical study (ChiCTR2100045380) [<span>5, 6</span>]. Here, we determined SYHA1813's antitumor activity in tumor models bearing <i>KRAS</i> or <i>EGFR</i> mutations.</p><p>We first examined the effects of SYHA1813 against cell line-derived xenograft (CDX) tumor models containing <i>KRAS<sup>G12C</sup></i> mutation (NCI-H358 lung cancer), <i>KRAS<sup>G12D</sup></i> mutation (PANC-1 pancreatic cancer) and wild-type <i>KRAS</i> (<i>KRAS<sup>WT</sup></i>) (HT-29 colorectal cancer). The results demonstrated that oral administration of SYHA1813 at a dose of 10 mg/kg significantly reduced tumor growth in the NCI-H358 xenograft model, with comparable efficacy to the US Food and Drug Administration (FDA) approved KRAS<sup>G12C</sup> inhibitor sotorasib (AMG510) (Figure 1A). SYHA1813 treatment also resulted in tumor regression in PANC-1 and HT-29 xenograft models (Figure 1B-C). No significant body weight loss was observed in all groups (Supplementary Figure S1). Moreover, considering the emergence of drug resistance as a significant challenge of AMG510, we established a drug resistance model of AMG510 (designated as AMG510R). We found that although AMG510 exhibited attenuated efficacy against the AMG510R model compared to the NCI-H358 model, SYHA1813 could still suppress the growth of drug-resistant tumors at the same dose (Figure 1D). Furthermore, SYHA1813 was evaluated in two patient-derived xenograft (PDX) models, including gastric tumor model GC-1-005 (<i>KRAS<sup>G13D</sup></i>) and colorect
癌基因是多种癌症类型肿瘤发生的关键因素,在肿瘤免疫逃逸中发挥着重要作用。Kirsten 大鼠肉瘤病毒癌基因同源物(KRAS)和表皮生长因子受体(EGFR)的突变是最常见的功能获得性改变[1]。经过多年的深入研究,针对表皮生长因子受体(EGFR)或 KRAS 突变的抑制剂已被成功开发出来,但其临床疗效相对有限,且不可避免地会遇到耐药性的挑战。耐药性的出现归因于驱动基因的二次突变和其他复杂因素。值得注意的是,对于不同类型的 KRAS 或 EGFR 突变的肿瘤,包括肿瘤中常见的 KRASG12D、KRASG13D 和 EGFRC797S 突变,目前尚缺乏获批的治疗策略[2]。此外,癌基因突变可能引发一连串的肿瘤微环境变化,最终导致肿瘤进展或对程序性死亡-1(PD-1)抗体疗法产生抗药性[3, 4]。SYHA1813是一种新型血管内皮生长因子受体(VEGFR)和集落刺激因子1受体(CSF1R)双重抑制剂,在临床前通过抑制血管生成和促进肿瘤免疫表现出强大的抗胶质瘤活性,并在一项正在进行的临床研究(ChiCTR2100045380)中显示出良好的疗效[5, 6]。我们首先研究了SYHA1813对含有KRASG12C突变(NCI-H358肺癌)、KRASG12D突变(PANC-1胰腺癌)和野生型KRAS(KRASWT)(HT-29结直肠癌)的细胞系衍生异种移植(CDX)肿瘤模型的作用。结果表明,在NCI-H358异种移植模型中,口服10毫克/千克剂量的SYHA1813可显著降低肿瘤生长,其疗效与美国食品药品管理局(FDA)批准的KRASG12C抑制剂索托拉西布(AMG510)相当(图1A)。SYHA1813治疗还导致PANC-1和HT-29异种移植模型中的肿瘤消退(图1B-C)。所有组均未观察到明显的体重下降(补充图 S1)。此外,考虑到耐药性的出现是 AMG510 面临的一个重大挑战,我们建立了 AMG510 的耐药模型(命名为 AMG510R)。我们发现,虽然与 NCI-H358 模型相比,AMG510 对 AMG510R 模型的疗效有所减弱,但 SYHA1813 仍能在相同剂量下抑制耐药肿瘤的生长(图 1D)。此外,SYHA1813还在两种患者来源异种移植(PDX)模型中进行了评估,包括胃肿瘤模型GC-1-005(KRASG13D)和结直肠肿瘤模型CRC-1-003(KRASWT)(图1E)。SYHA1813 在这些 PDX 模型中也表现出了强大的抗肿瘤活性(图 1F-G)。鉴于 SYHA1813 通过调节血管生成和巨噬细胞发挥抗肿瘤作用,我们进一步分析了 NCI-H358 和 HT-29 肿瘤组织中与血管生成、巨噬细胞和增殖相关的标记物的表达。免疫组化(IHC)结果显示,SYHA1813能显著降低血管生成标志物CD31、巨噬细胞标志物F4/80、M2型巨噬细胞标志物CD206和精氨酸酶-1(ARG1)以及肿瘤增殖标志物Ki67的表达(图1H和补充图S2)。总之,SYHA1813在一组基因和组织学异质性的KRAS突变肿瘤模型(包括对AMG510耐药的模型)中表现出强大的抗肿瘤活性。为了解决这个问题,我们首先采用了含有EGFRL858R/T790M突变的NCI-H1975模型,该模型对第一代表皮生长因子受体TKIs耐药,结果发现SYHA1813能显著抑制肿瘤的生长(图1I)。然后,我们评估了 SYHA1813 对携带 EGFRC797S 三重突变的肿瘤模型(PC-9-OR)的活性,该模型对第三代 EGFR TKIs 具有耐药性[7]。如图 1J 所示,美国 FDA 批准的第一种第三代 EGFR TKI--奥西替尼(AZD9291)即使剂量为 10 mg/kg,也无法抑制肿瘤生长。然而,我们观察到 SYHA1813 在 5 毫克/公斤和 10 毫克/公斤剂量下的肿瘤生长速度明显减慢(图 1J)。此外,我们还采用了奥希替尼耐药肿瘤(命名为 AZDR)[8],也观察到 SYHA1813 单药治疗后肿瘤生长速度明显下降(图 1K)。同样,在另一个对第三代表皮生长因子受体 TKI ASK120067 [8]耐药的模型(命名为 67R)中,SYHA1813 单药治疗也抑制了肿瘤生长(图 1L)。IHC 结果还显示,服用 SYHA1813 后,肿瘤组织中的血管生成标记物和巨噬细胞标记物减少(补充图 S3)。
{"title":"A novel strategy for treating oncogene-mutated tumors by targeting tumor microenvironment and synergistically enhancing anti-PD-1 immunotherapy","authors":"Yingqiang Liu,&nbsp;Linjiang Tong,&nbsp;Mengge Zhang,&nbsp;Qi Zhang,&nbsp;Qiupei Liu,&nbsp;Fang Feng,&nbsp;Yan Li,&nbsp;Mengzhen Lai,&nbsp;Haotian Tang,&nbsp;Yi Chen,&nbsp;Meiyu Geng,&nbsp;Wenhu Duan,&nbsp;Jian Ding,&nbsp;Hua Xie","doi":"10.1002/cac2.12521","DOIUrl":"10.1002/cac2.12521","url":null,"abstract":"&lt;p&gt;Oncogenes are critical factors in tumorigenesis of diverse cancer types and play essential roles in tumor immune escape. Mutations in Kirsten rat sarcoma viral oncogene homolog (&lt;i&gt;KRAS&lt;/i&gt;) and epidermal growth factor receptor (&lt;i&gt;EGFR&lt;/i&gt;) are among the most frequent gain-of-function alterations [&lt;span&gt;1&lt;/span&gt;]. After many years of in-depth research, inhibitors targeting &lt;i&gt;EGFR&lt;/i&gt; or &lt;i&gt;KRAS&lt;/i&gt; mutations have been successfully developed, however, their clinical benefit is relatively limited, and they will inevitably encounter the challenge of drug resistance. The emergence of resistance is attributed to secondary mutations in driver genes and other complicated factors. It is worth noting that approved treatment strategies are currently lacking for tumors with different types of &lt;i&gt;KRAS&lt;/i&gt; or &lt;i&gt;EGFR&lt;/i&gt; mutations, including &lt;i&gt;KRAS&lt;sup&gt;G12D&lt;/sup&gt;&lt;/i&gt;, &lt;i&gt;KRAS&lt;sup&gt;G13D&lt;/sup&gt;&lt;/i&gt;, and &lt;i&gt;EGFR&lt;sup&gt;C797S&lt;/sup&gt;&lt;/i&gt; mutations that are common in tumors [&lt;span&gt;2&lt;/span&gt;]. Additionally, oncogene mutations could trigger a cascade of tumor microenvironment changes, ultimately resulting in tumor progression or resistance to programmed death-1 (PD-1) antibody therapy [&lt;span&gt;3, 4&lt;/span&gt;]. SYHA1813, a novel vascular endothelial growth factor receptor (VEGFR) and colony-stimulating factor 1 receptor (CSF1R) dual inhibitor, exhibited potent preclinical anti-glioma activity by inhibiting angiogenesis and promoting tumor immunity and showed promising efficacy in an ongoing clinical study (ChiCTR2100045380) [&lt;span&gt;5, 6&lt;/span&gt;]. Here, we determined SYHA1813's antitumor activity in tumor models bearing &lt;i&gt;KRAS&lt;/i&gt; or &lt;i&gt;EGFR&lt;/i&gt; mutations.&lt;/p&gt;&lt;p&gt;We first examined the effects of SYHA1813 against cell line-derived xenograft (CDX) tumor models containing &lt;i&gt;KRAS&lt;sup&gt;G12C&lt;/sup&gt;&lt;/i&gt; mutation (NCI-H358 lung cancer), &lt;i&gt;KRAS&lt;sup&gt;G12D&lt;/sup&gt;&lt;/i&gt; mutation (PANC-1 pancreatic cancer) and wild-type &lt;i&gt;KRAS&lt;/i&gt; (&lt;i&gt;KRAS&lt;sup&gt;WT&lt;/sup&gt;&lt;/i&gt;) (HT-29 colorectal cancer). The results demonstrated that oral administration of SYHA1813 at a dose of 10 mg/kg significantly reduced tumor growth in the NCI-H358 xenograft model, with comparable efficacy to the US Food and Drug Administration (FDA) approved KRAS&lt;sup&gt;G12C&lt;/sup&gt; inhibitor sotorasib (AMG510) (Figure 1A). SYHA1813 treatment also resulted in tumor regression in PANC-1 and HT-29 xenograft models (Figure 1B-C). No significant body weight loss was observed in all groups (Supplementary Figure S1). Moreover, considering the emergence of drug resistance as a significant challenge of AMG510, we established a drug resistance model of AMG510 (designated as AMG510R). We found that although AMG510 exhibited attenuated efficacy against the AMG510R model compared to the NCI-H358 model, SYHA1813 could still suppress the growth of drug-resistant tumors at the same dose (Figure 1D). Furthermore, SYHA1813 was evaluated in two patient-derived xenograft (PDX) models, including gastric tumor model GC-1-005 (&lt;i&gt;KRAS&lt;sup&gt;G13D&lt;/sup&gt;&lt;/i&gt;) and colorect","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":"44 3","pages":"438-442"},"PeriodicalIF":16.2,"publicationDate":"2024-02-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12521","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139711572","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
Cover Image, Volume 44, Issue 1 封面图片,第 44 卷第 1 期
IF 16.2 1区 医学 Q1 ONCOLOGY Pub Date : 2024-02-06 DOI: 10.1002/cac2.12522
Monika Raab, Izabela Kostova, Samuel Peña-Llopis, Daniela Fietz, Monika Kressin, Seyed Mohsen Aberoumandi, Evelyn Ullrich, Sven Becker, Mourad Sanhaji, Klaus Strebhardt

The cover image is based on the Original Article Rescue of p53 functions by in vitro-transcribed mRNA impedes the growth of highgrade serous ovarian cancer by Monika Raab et al., https://doi.org/10.1002/cac2.12511.

封面图片基于 Monika Raab 等人的原创文章《体外转录 mRNA 挽救 p53 功能阻碍高级别浆液性卵巢癌的生长》,https://doi.org/10.1002/cac2.12511。
{"title":"Cover Image, Volume 44, Issue 1","authors":"Monika Raab,&nbsp;Izabela Kostova,&nbsp;Samuel Peña-Llopis,&nbsp;Daniela Fietz,&nbsp;Monika Kressin,&nbsp;Seyed Mohsen Aberoumandi,&nbsp;Evelyn Ullrich,&nbsp;Sven Becker,&nbsp;Mourad Sanhaji,&nbsp;Klaus Strebhardt","doi":"10.1002/cac2.12522","DOIUrl":"https://doi.org/10.1002/cac2.12522","url":null,"abstract":"<p>The cover image is based on the Original Article <i>Rescue of p53 functions by in vitro-transcribed mRNA impedes the growth of highgrade serous ovarian cancer</i> by Monika Raab et al., https://doi.org/10.1002/cac2.12511.\u0000\u0000 <figure>\u0000 <div><picture>\u0000 <source></source></picture><p></p>\u0000 </div>\u0000 </figure></p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":"44 1","pages":""},"PeriodicalIF":16.2,"publicationDate":"2024-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12522","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139695464","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
Cell fate regulation governed by p53: Friends or reversible foes in cancer therapy 受 p53 调控的细胞命运:癌症治疗中的朋友还是可逆的敌人
IF 16.2 1区 医学 Q1 ONCOLOGY Pub Date : 2024-02-04 DOI: 10.1002/cac2.12520
Bin Song, Ping Yang, Shuyu Zhang

Cancer is a leading cause of death worldwide. Targeted therapies aimed at key oncogenic driver mutations in combination with chemotherapy and radiotherapy as well as immunotherapy have benefited cancer patients considerably. Tumor protein p53 (TP53), a crucial tumor suppressor gene encoding p53, regulates numerous downstream genes and cellular phenotypes in response to various stressors. The affected genes are involved in diverse processes, including cell cycle arrest, DNA repair, cellular senescence, metabolic homeostasis, apoptosis, and autophagy. However, accumulating recent studies have continued to reveal novel and unexpected functions of p53 in governing the fate of tumors, for example, functions in ferroptosis, immunity, the tumor microenvironment and microbiome metabolism. Among the possibilities, the evolutionary plasticity of p53 is the most controversial, partially due to the dizzying array of biological functions that have been attributed to different regulatory mechanisms of p53 signaling. Nearly 40 years after its discovery, this key tumor suppressor remains somewhat enigmatic. The intricate and diverse functions of p53 in regulating cell fate during cancer treatment are only the tip of the iceberg with respect to its equally complicated structural biology, which has been painstakingly revealed. Additionally, TP53 mutation is one of the most significant genetic alterations in cancer, contributing to rapid cancer cell growth and tumor progression. Here, we summarized recent advances that implicate altered p53 in modulating the response to various cancer therapies, including chemotherapy, radiotherapy, and immunotherapy. Furthermore, we also discussed potential strategies for targeting p53 as a therapeutic option for cancer.

癌症是导致全球死亡的主要原因。针对关键致癌驱动基因突变的靶向疗法与化疗、放疗和免疫疗法相结合,使癌症患者受益匪浅。肿瘤蛋白 p53(TP53)是一种重要的肿瘤抑制基因,编码 p53,可调节众多下游基因和细胞表型,以应对各种压力。受影响的基因参与多种过程,包括细胞周期停滞、DNA 修复、细胞衰老、代谢平衡、细胞凋亡和自噬。然而,最近不断积累的研究继续揭示了 p53 在调控肿瘤命运方面的新功能和意想不到的功能,例如在铁变态反应、免疫、肿瘤微环境和微生物组代谢方面的功能。在这些可能性中,p53 的进化可塑性最具争议性,部分原因是 p53 信号的不同调控机制具有令人眼花缭乱的生物功能。在发现 p53 近 40 年后,这个关键的肿瘤抑制因子仍然是个谜。p53 在癌症治疗过程中调节细胞命运的功能错综复杂、多种多样,但这仅仅是冰山一角,其同样复杂的结构生物学特性已被煞费苦心地揭示出来。此外,TP53 基因突变是癌症中最重要的基因改变之一,导致癌细胞快速生长和肿瘤进展。在此,我们总结了最近的研究进展,这些进展表明,p53 的改变与调节对各种癌症疗法(包括化疗、放疗和免疫疗法)的反应有关。此外,我们还讨论了将 p53 作为癌症治疗靶点的潜在策略。
{"title":"Cell fate regulation governed by p53: Friends or reversible foes in cancer therapy","authors":"Bin Song,&nbsp;Ping Yang,&nbsp;Shuyu Zhang","doi":"10.1002/cac2.12520","DOIUrl":"10.1002/cac2.12520","url":null,"abstract":"<p>Cancer is a leading cause of death worldwide. Targeted therapies aimed at key oncogenic driver mutations in combination with chemotherapy and radiotherapy as well as immunotherapy have benefited cancer patients considerably. Tumor protein p53 (<i>TP53</i>), a crucial tumor suppressor gene encoding p53, regulates numerous downstream genes and cellular phenotypes in response to various stressors. The affected genes are involved in diverse processes, including cell cycle arrest, DNA repair, cellular senescence, metabolic homeostasis, apoptosis, and autophagy. However, accumulating recent studies have continued to reveal novel and unexpected functions of p53 in governing the fate of tumors, for example, functions in ferroptosis, immunity, the tumor microenvironment and microbiome metabolism. Among the possibilities, the evolutionary plasticity of p53 is the most controversial, partially due to the dizzying array of biological functions that have been attributed to different regulatory mechanisms of p53 signaling. Nearly 40 years after its discovery, this key tumor suppressor remains somewhat enigmatic. The intricate and diverse functions of p53 in regulating cell fate during cancer treatment are only the tip of the iceberg with respect to its equally complicated structural biology, which has been painstakingly revealed. Additionally, <i>TP53</i> mutation is one of the most significant genetic alterations in cancer, contributing to rapid cancer cell growth and tumor progression. Here, we summarized recent advances that implicate altered p53 in modulating the response to various cancer therapies, including chemotherapy, radiotherapy, and immunotherapy. Furthermore, we also discussed potential strategies for targeting p53 as a therapeutic option for cancer.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":"44 3","pages":"297-360"},"PeriodicalIF":16.2,"publicationDate":"2024-02-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12520","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139689067","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
AP4 induces JNK1 and a miR-22-3p/FOSL1 feed-forward loop to activate AP-1 and promote colorectal cancer metastasis AP4 可诱导 JNK1 和 miR-22-3p/FOSL1 前馈环,从而激活 AP-1,促进结直肠癌转移。
IF 16.2 1区 医学 Q1 ONCOLOGY Pub Date : 2024-01-15 DOI: 10.1002/cac2.12514
Jinjiang Chou, Markus Kaller, Matjaz Rokavec, Fangteng Liu, Heiko Hermeking
<p>Dear Editor,</p><p>Colorectal cancer (CRC) is the third most deadly cancer worldwide [<span>1</span>]. The mortality of CRC has remained high due to limited treatment options for metastatic CRC (mCRC) [<span>2</span>]. Epithelial-mesenchymal transition (EMT) is an important contributor to mCRC [<span>2</span>]. The c-MYC proto-oncogene (MYC)-induced transcription factor AP4 (TFAP4/AP4) is a driver of EMT, thereby presumably facilitates mCRC [<span>3, 4</span>]. The mitogen-activated protein kinase (MAPK)/c-Jun N-terminal kinase (JNK)/activator protein-1 (AP-1) pathway has been implicated in the regulation of EMT and mCRC [<span>5</span>].</p><p>Here, we analyzed whether AP4 regulates components of the MAPK/JNK/AP-1 pathway after MYC activation using CRC cells rendered <i>AP4</i>-deficient by a clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) approach. The detailed methods are shown in the Supplementary file. First, we grouped MYC-induced changes in mRNA expression observed in the CRC cell line DLD-1 <i>AP4</i> wild-type 1/pRTR-<i>c-MYC</i>-VSV (<i>AP4</i>-WT1 DLD-1/pRTR-<i>c</i>-<i>MYC-</i>VSV) into 6 non-overlapping expression clusters (Supplementary Figure S1A, left), with cluster 1 representing mRNAs down-regulated, and clusters 2-6 representing different patterns of mRNA up-regulated after MYC activation. MAPK signaling pathway components were strongly over-represented in cluster 6 (Supplementary Figure S1A, right). The AP4 targets <i>MIR22 host gene</i> (<i>MIR22HG</i>) and <i>E-</i><i>cadherin 1</i> (<i>CDH1</i>) were down-regulated after MYC activation in <i>AP4</i>-WT1 DLD-1/pRTR-<i>c-MYC</i>-VSV cells (Supplementary Figure S1B). MAPK signaling effectors, including c-<i>Fos proto-oncogene</i> (<i>FOS</i>), c-<i>Jun proto-oncogene</i> (<i>JUN</i>) and <i>c-J</i><i>unB proto-oncogene</i> (<i>JUNB</i>), were over-represented in cluster 6. Additional MAPK signaling pathway components. such as <i>c-Jun N-terminal kinase 1</i> (<i>JNK1</i>), <i>mitogen-activated protein kinase kinase kinase 1</i> (<i>MAP3K1</i>), <i>mitogen-activated protein kinase kinase kinase 13</i> (<i>MAP3K13</i>), <i>mitogen-activated protein kinase kinase 3</i> (<i>MAP2K3</i>), <i>mitogen-activated protein kinase kinase 7</i> (<i>MAP2K7</i>) and <i>FOS</i>-<i>like</i> <i>1</i> (<i>FOSL1</i>) were found in clusters 3-5 (Supplementary Figure S1B). Interestingly, <i>MAP3K13, MAP2K7, JNK1</i> and <i>FOSL1</i> were induced by MYC in an <i>AP4</i>-dependent manner (Supplementary Figure S1C-D).</p><p>Notably, <i>MAP3K13</i>, <i>FOSL1, JNK1</i> and <i>MAP2K7</i> were also up-regulated after activating <i>AP4</i> for 48 or 72 hours (Figure 1A) and showed AP4-binding sites (CAGCTG) and AP4 occupancy (Supplementary Figure S2A). Therefore, these genes presumably represent direct AP4 targets. MAP3K13 and FOSL1 protein and phosphorylated JNK1 were up-regulated after <i>AP4</i> activation, whereas JNK1 protein levels remained unch
由于我们在 FOSL1 3'- 非翻译区(3'-UTR)发现了 miR-22-3p 种子匹配序列(SMS),因此我们研究了 FOSL1 是否也通过抑制 miR-22-3p 受 AP4 的调控(图 1F)。事实上,FOSL1 3'-UTR 报告受异位 miR-22-3p 的抑制,而 SMS 的突变使报告对 miR-22-3p 难以承受(图 1G)。异位 miR-22-3p 抑制了 FOSL1(补充图 S4A),并有效降低了基础和 AP4 诱导的 FOSL1 蛋白水平(图 1H,补充图 S4B)。接下来,我们生成了 MIR22-/- (MIR22-基因剔除 [KO])和 FOSL1 ΔSMS CRC 细胞系(补充图 S4C)。在 MIR22 缺失的 CRC 细胞中,FOSL1 mRNA 和蛋白表达均升高(图 1I,补图 S4D)。MIR22 缺失以 miR-22-3p SMS 依赖性方式增加了 FOSL1 3'-UTR 报告的活性(补充图 S4E),并增强了迁移和侵袭(补充图 S4F)。此外,在 MIR22 缺失的 CRC 细胞中,固有 AP-1 活性显著升高(图 1J,补图 S4G)。MIR22 缺陷和 FOSL1 ΔSMS DLD-1 细胞中 FOSL1 mRNA 的半衰期(t1/2)延长(图 1K)。在 FOSL1 ΔSMS CRC 细胞中异位表达 miR-22-3p 未能抑制 FOSL1(图 1L,补充图 S4H),这表明 FOSL1 的基础表达增加,并减弱了 AP4 对 FOSL1 的上调(图 1M)。因此,miR-22-3p对FOSL1的直接靶向作用对于维持低水平的FOSL1表达至关重要,这使得FOSL1可以被AP4直接诱导,并通过抑制MIR22间接诱导。异位 AP4 可抑制 CDH1 并诱导 CRC 细胞中的 VIM,而沉默 FOSL1 则会部分削弱这些效应(补充图 S5A)。异位AP4对迁移和侵袭的积极作用在很大程度上被FOSL1沉默所削弱(图1N,补充图S5B-C),这表明FOSL1介导了AP4的这些作用。沉默 FOSL1 能显著抑制 AP-1 活性,并削弱 AP4 对 AP-1 活性的影响(补充图 S5D)。此外,异位 AP4 会诱导 DLD-1 细胞增殖,而同时 siRNA 介导的 FOSL1 沉默会抑制这种增殖(补充图 S5E)。综上所述,这些结果表明,AP4对EMT、迁移、侵袭和增殖的影响需要FOSL1介导的AP-1激活。因此,AP4对miR-22-3p/FOSL1轴的调控可能与mCRC的临床相关。值得注意的是,通过非侵入性生物发光成像(图 1O-P)和组织学分析(补充图 S6A,右图)确定,非转移性 DLD-1 细胞中的 AP4 激活可使这些细胞在非肥胖糖尿病/严重联合免疫缺陷(NOD/SCID)小鼠体内形成肺转移。因此,AP4 的激活足以使 CRC 细胞从非转移状态转变为转移状态。沉默 FOSL1 能有效阻止 AP4 诱导的肺转移的形成(图 1Q,补充图 S6A 左)。因此,FOSL1介导了AP4诱导的转移的形成,并且是形成转移所必需的。使用 DB07268 抑制 JNK1 后也得到了类似的结果(图 1R-S,补充图 S6B-C)。AP4、miR-22-3p 和 FOSL1 形成了一个连贯的前馈调节环。这些调控汇聚在 AP-1 的激活上(图 1T),最终促进了 EMT、迁移和侵袭,从而加强了转移的形成。Heiko Hermeking构思、规划并指导了该项目;Heiko Hermeking、周锦江和Markus Kaller设计了实验;周锦江进行了实验并分析了结果;Markus Kaller进行了生物信息学分析;Matjaz Rokavec进行了小鼠实验并分析了结果。刘方腾对小鼠肺转移进行了组织化学评估。Heiko Hermeking、周锦江和Markus Kaller撰写了手稿。动物实验和分析获得了德国上巴伐利亚州政府的批准(55.2-2532.vet_02-18-57)。
{"title":"AP4 induces JNK1 and a miR-22-3p/FOSL1 feed-forward loop to activate AP-1 and promote colorectal cancer metastasis","authors":"Jinjiang Chou,&nbsp;Markus Kaller,&nbsp;Matjaz Rokavec,&nbsp;Fangteng Liu,&nbsp;Heiko Hermeking","doi":"10.1002/cac2.12514","DOIUrl":"10.1002/cac2.12514","url":null,"abstract":"&lt;p&gt;Dear Editor,&lt;/p&gt;&lt;p&gt;Colorectal cancer (CRC) is the third most deadly cancer worldwide [&lt;span&gt;1&lt;/span&gt;]. The mortality of CRC has remained high due to limited treatment options for metastatic CRC (mCRC) [&lt;span&gt;2&lt;/span&gt;]. Epithelial-mesenchymal transition (EMT) is an important contributor to mCRC [&lt;span&gt;2&lt;/span&gt;]. The c-MYC proto-oncogene (MYC)-induced transcription factor AP4 (TFAP4/AP4) is a driver of EMT, thereby presumably facilitates mCRC [&lt;span&gt;3, 4&lt;/span&gt;]. The mitogen-activated protein kinase (MAPK)/c-Jun N-terminal kinase (JNK)/activator protein-1 (AP-1) pathway has been implicated in the regulation of EMT and mCRC [&lt;span&gt;5&lt;/span&gt;].&lt;/p&gt;&lt;p&gt;Here, we analyzed whether AP4 regulates components of the MAPK/JNK/AP-1 pathway after MYC activation using CRC cells rendered &lt;i&gt;AP4&lt;/i&gt;-deficient by a clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) approach. The detailed methods are shown in the Supplementary file. First, we grouped MYC-induced changes in mRNA expression observed in the CRC cell line DLD-1 &lt;i&gt;AP4&lt;/i&gt; wild-type 1/pRTR-&lt;i&gt;c-MYC&lt;/i&gt;-VSV (&lt;i&gt;AP4&lt;/i&gt;-WT1 DLD-1/pRTR-&lt;i&gt;c&lt;/i&gt;-&lt;i&gt;MYC-&lt;/i&gt;VSV) into 6 non-overlapping expression clusters (Supplementary Figure S1A, left), with cluster 1 representing mRNAs down-regulated, and clusters 2-6 representing different patterns of mRNA up-regulated after MYC activation. MAPK signaling pathway components were strongly over-represented in cluster 6 (Supplementary Figure S1A, right). The AP4 targets &lt;i&gt;MIR22 host gene&lt;/i&gt; (&lt;i&gt;MIR22HG&lt;/i&gt;) and &lt;i&gt;E-&lt;/i&gt;&lt;i&gt;cadherin 1&lt;/i&gt; (&lt;i&gt;CDH1&lt;/i&gt;) were down-regulated after MYC activation in &lt;i&gt;AP4&lt;/i&gt;-WT1 DLD-1/pRTR-&lt;i&gt;c-MYC&lt;/i&gt;-VSV cells (Supplementary Figure S1B). MAPK signaling effectors, including c-&lt;i&gt;Fos proto-oncogene&lt;/i&gt; (&lt;i&gt;FOS&lt;/i&gt;), c-&lt;i&gt;Jun proto-oncogene&lt;/i&gt; (&lt;i&gt;JUN&lt;/i&gt;) and &lt;i&gt;c-J&lt;/i&gt;&lt;i&gt;unB proto-oncogene&lt;/i&gt; (&lt;i&gt;JUNB&lt;/i&gt;), were over-represented in cluster 6. Additional MAPK signaling pathway components. such as &lt;i&gt;c-Jun N-terminal kinase 1&lt;/i&gt; (&lt;i&gt;JNK1&lt;/i&gt;), &lt;i&gt;mitogen-activated protein kinase kinase kinase 1&lt;/i&gt; (&lt;i&gt;MAP3K1&lt;/i&gt;), &lt;i&gt;mitogen-activated protein kinase kinase kinase 13&lt;/i&gt; (&lt;i&gt;MAP3K13&lt;/i&gt;), &lt;i&gt;mitogen-activated protein kinase kinase 3&lt;/i&gt; (&lt;i&gt;MAP2K3&lt;/i&gt;), &lt;i&gt;mitogen-activated protein kinase kinase 7&lt;/i&gt; (&lt;i&gt;MAP2K7&lt;/i&gt;) and &lt;i&gt;FOS&lt;/i&gt;-&lt;i&gt;like&lt;/i&gt; &lt;i&gt;1&lt;/i&gt; (&lt;i&gt;FOSL1&lt;/i&gt;) were found in clusters 3-5 (Supplementary Figure S1B). Interestingly, &lt;i&gt;MAP3K13, MAP2K7, JNK1&lt;/i&gt; and &lt;i&gt;FOSL1&lt;/i&gt; were induced by MYC in an &lt;i&gt;AP4&lt;/i&gt;-dependent manner (Supplementary Figure S1C-D).&lt;/p&gt;&lt;p&gt;Notably, &lt;i&gt;MAP3K13&lt;/i&gt;, &lt;i&gt;FOSL1, JNK1&lt;/i&gt; and &lt;i&gt;MAP2K7&lt;/i&gt; were also up-regulated after activating &lt;i&gt;AP4&lt;/i&gt; for 48 or 72 hours (Figure 1A) and showed AP4-binding sites (CAGCTG) and AP4 occupancy (Supplementary Figure S2A). Therefore, these genes presumably represent direct AP4 targets. MAP3K13 and FOSL1 protein and phosphorylated JNK1 were up-regulated after &lt;i&gt;AP4&lt;/i&gt; activation, whereas JNK1 protein levels remained unch","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":"44 3","pages":"433-437"},"PeriodicalIF":16.2,"publicationDate":"2024-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12514","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139472306","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
Cellular metabolism: A key player in cancer ferroptosis 细胞新陈代谢:癌症铁变态反应的关键角色
IF 16.2 1区 医学 Q1 ONCOLOGY Pub Date : 2024-01-13 DOI: 10.1002/cac2.12519
Xianjie Jiang, Qiu Peng, Mingjing Peng, Linda Oyang, Honghan Wang, Qiang Liu, Xuemeng Xu, Nayiyuan Wu, Shiming Tan, Wenjuan Yang, Yaqian Han, Jinguan Lin, Longzheng Xia, Yanyan Tang, Xia Luo, Jie Dai, Yujuan Zhou, Qianjin Liao

Cellular metabolism is the fundamental process by which cells maintain growth and self-renewal. It produces energy, furnishes raw materials, and intermediates for biomolecule synthesis, and modulates enzyme activity to sustain normal cellular functions. Cellular metabolism is the foundation of cellular life processes and plays a regulatory role in various biological functions, including programmed cell death. Ferroptosis is a recently discovered form of iron-dependent programmed cell death. The inhibition of ferroptosis plays a crucial role in tumorigenesis and tumor progression. However, the role of cellular metabolism, particularly glucose and amino acid metabolism, in cancer ferroptosis is not well understood. Here, we reviewed glucose, lipid, amino acid, iron and selenium metabolism involvement in cancer cell ferroptosis to elucidate the impact of different metabolic pathways on this process. Additionally, we provided a detailed overview of agents used to induce cancer ferroptosis. We explained that the metabolism of tumor cells plays a crucial role in maintaining intracellular redox homeostasis and that disrupting the normal metabolic processes in these cells renders them more susceptible to iron-induced cell death, resulting in enhanced tumor cell killing. The combination of ferroptosis inducers and cellular metabolism inhibitors may be a novel approach to future cancer therapy and an important strategy to advance the development of treatments.

细胞新陈代谢是细胞维持生长和自我更新的基本过程。它产生能量,为生物大分子合成提供原料和中间体,并调节酶的活性以维持正常的细胞功能。细胞新陈代谢是细胞生命过程的基础,在包括细胞程序性死亡在内的各种生物功能中发挥着调节作用。铁突变是最近发现的一种依赖铁的细胞程序性死亡形式。抑制铁跃迁在肿瘤发生和发展中起着至关重要的作用。然而,人们对细胞代谢,尤其是葡萄糖和氨基酸代谢在癌症铁凋亡中的作用还不甚了解。在此,我们回顾了葡萄糖、脂质、氨基酸、铁和硒代谢在癌细胞铁跃迁中的参与,以阐明不同代谢途径对这一过程的影响。此外,我们还详细介绍了用于诱导癌细胞铁变态反应的药物。我们解释说,肿瘤细胞的新陈代谢在维持细胞内氧化还原平衡方面起着至关重要的作用,破坏这些细胞的正常新陈代谢过程会使它们更容易受到铁诱导的细胞死亡的影响,从而增强对肿瘤细胞的杀伤力。铁氧化诱导剂和细胞代谢抑制剂的结合可能是未来癌症治疗的一种新方法,也是推动治疗方法发展的一种重要策略。
{"title":"Cellular metabolism: A key player in cancer ferroptosis","authors":"Xianjie Jiang,&nbsp;Qiu Peng,&nbsp;Mingjing Peng,&nbsp;Linda Oyang,&nbsp;Honghan Wang,&nbsp;Qiang Liu,&nbsp;Xuemeng Xu,&nbsp;Nayiyuan Wu,&nbsp;Shiming Tan,&nbsp;Wenjuan Yang,&nbsp;Yaqian Han,&nbsp;Jinguan Lin,&nbsp;Longzheng Xia,&nbsp;Yanyan Tang,&nbsp;Xia Luo,&nbsp;Jie Dai,&nbsp;Yujuan Zhou,&nbsp;Qianjin Liao","doi":"10.1002/cac2.12519","DOIUrl":"10.1002/cac2.12519","url":null,"abstract":"<p>Cellular metabolism is the fundamental process by which cells maintain growth and self-renewal. It produces energy, furnishes raw materials, and intermediates for biomolecule synthesis, and modulates enzyme activity to sustain normal cellular functions. Cellular metabolism is the foundation of cellular life processes and plays a regulatory role in various biological functions, including programmed cell death. Ferroptosis is a recently discovered form of iron-dependent programmed cell death. The inhibition of ferroptosis plays a crucial role in tumorigenesis and tumor progression. However, the role of cellular metabolism, particularly glucose and amino acid metabolism, in cancer ferroptosis is not well understood. Here, we reviewed glucose, lipid, amino acid, iron and selenium metabolism involvement in cancer cell ferroptosis to elucidate the impact of different metabolic pathways on this process. Additionally, we provided a detailed overview of agents used to induce cancer ferroptosis. We explained that the metabolism of tumor cells plays a crucial role in maintaining intracellular redox homeostasis and that disrupting the normal metabolic processes in these cells renders them more susceptible to iron-induced cell death, resulting in enhanced tumor cell killing. The combination of ferroptosis inducers and cellular metabolism inhibitors may be a novel approach to future cancer therapy and an important strategy to advance the development of treatments.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":"44 2","pages":"185-204"},"PeriodicalIF":16.2,"publicationDate":"2024-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12519","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139459766","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
The Chinese Society of Clinical Oncology (CSCO): Clinical guidelines for the diagnosis and treatment of gastric cancer, 2023 中国临床肿瘤学会(CSCO):2023 年胃癌诊断与治疗临床指南
IF 16.2 1区 医学 Q1 ONCOLOGY Pub Date : 2023-12-31 DOI: 10.1002/cac2.12516
Feng-Hua Wang, Xiao-Tian Zhang, Lei Tang, Qi Wu, Mu-Yan Cai, Yuan-Fang Li, Xiu-Juan Qu, Hong Qiu, Yu-Jing Zhang, Jie-Er Ying, Jun Zhang, Ling-Yu Sun, Rong-Bo Lin, Chang Wang, Hao Liu, Miao-Zhen Qiu, Wen-Long Guan, Sheng-Xiang Rao, Jia-Fu Ji, Yan Xin, Wei-Qi Sheng, Hui-Mian Xu, Zhi-Wei Zhou, Ai-Ping Zhou, Jing Jin, Xiang-Lin Yuan, Feng Bi, Tian-Shu Liu, Han Liang, Yan-Qiao Zhang, Guo-Xin Li, Jun Liang, Bao-Rui Liu, Lin Shen, Jin Li, Rui-Hua Xu

The 2023 update of the Chinese Society of Clinical Oncology (CSCO) Clinical Guidelines for Gastric Cancer focuses on standardizing cancer diagnosis and treatment in China, reflecting the latest advancements in evidence-based medicine, healthcare resource availability, and precision medicine. These updates address the differences in epidemiological characteristics, clinicopathological features, tumor biology, treatment patterns, and drug selections between Eastern and Western gastric cancer patients. Key revisions include a structured template for imaging diagnosis reports, updated standards for molecular marker testing in pathological diagnosis, and an elevated recommendation for neoadjuvant chemotherapy in stage III gastric cancer. For advanced metastatic gastric cancer, the guidelines introduce new recommendations for immunotherapy, anti-angiogenic therapy and targeted drugs, along with updated management strategies for human epidermal growth factor receptor 2 (HER2)-positive and deficient DNA mismatch repair (dMMR)/microsatellite instability-high (MSI-H) patients. Additionally, the guidelines offer detailed screening recommendations for hereditary gastric cancer and an appendix listing drug treatment regimens for various stages of gastric cancer. The 2023 CSCO Clinical Guidelines for Gastric Cancer updates are based on both Chinese and international clinical research and expert consensus to enhance their applicability and relevance in clinical practice, particularly in the heterogeneous healthcare landscape of China, while maintaining a commitment to scientific rigor, impartiality, and timely revisions.

中国临床肿瘤学会(CSCO)《胃癌临床指南》2023年更新版致力于规范中国的癌症诊断和治疗,反映了循证医学、医疗资源可用性和精准医学的最新进展。这些更新解决了东西方胃癌患者在流行病学特征、临床病理特征、肿瘤生物学、治疗模式和药物选择方面的差异。主要修订内容包括影像诊断报告的结构化模板、病理诊断中分子标记物检测的最新标准,以及提高了对 III 期胃癌新辅助化疗的建议。对于晚期转移性胃癌,指南提出了免疫疗法、抗血管生成疗法和靶向药物的新建议,并更新了人表皮生长因子受体2(HER2)阳性和DNA错配修复缺陷(dMMR)/微卫星不稳定性高(MSI-H)患者的管理策略。此外,指南还提供了详细的遗传性胃癌筛查建议,并在附录中列出了不同阶段胃癌的药物治疗方案。2023年CSCO胃癌临床指南》的更新基于中外临床研究和专家共识,以提高其在临床实践中的适用性和相关性,尤其是在中国异质化的医疗环境中,同时保持科学严谨、公正和及时修订的承诺。
{"title":"The Chinese Society of Clinical Oncology (CSCO): Clinical guidelines for the diagnosis and treatment of gastric cancer, 2023","authors":"Feng-Hua Wang,&nbsp;Xiao-Tian Zhang,&nbsp;Lei Tang,&nbsp;Qi Wu,&nbsp;Mu-Yan Cai,&nbsp;Yuan-Fang Li,&nbsp;Xiu-Juan Qu,&nbsp;Hong Qiu,&nbsp;Yu-Jing Zhang,&nbsp;Jie-Er Ying,&nbsp;Jun Zhang,&nbsp;Ling-Yu Sun,&nbsp;Rong-Bo Lin,&nbsp;Chang Wang,&nbsp;Hao Liu,&nbsp;Miao-Zhen Qiu,&nbsp;Wen-Long Guan,&nbsp;Sheng-Xiang Rao,&nbsp;Jia-Fu Ji,&nbsp;Yan Xin,&nbsp;Wei-Qi Sheng,&nbsp;Hui-Mian Xu,&nbsp;Zhi-Wei Zhou,&nbsp;Ai-Ping Zhou,&nbsp;Jing Jin,&nbsp;Xiang-Lin Yuan,&nbsp;Feng Bi,&nbsp;Tian-Shu Liu,&nbsp;Han Liang,&nbsp;Yan-Qiao Zhang,&nbsp;Guo-Xin Li,&nbsp;Jun Liang,&nbsp;Bao-Rui Liu,&nbsp;Lin Shen,&nbsp;Jin Li,&nbsp;Rui-Hua Xu","doi":"10.1002/cac2.12516","DOIUrl":"10.1002/cac2.12516","url":null,"abstract":"<div>\u0000 \u0000 \u0000 <section>\u0000 \u0000 <p>The 2023 update of the Chinese Society of Clinical Oncology (CSCO) Clinical Guidelines for Gastric Cancer focuses on standardizing cancer diagnosis and treatment in China, reflecting the latest advancements in evidence-based medicine, healthcare resource availability, and precision medicine. These updates address the differences in epidemiological characteristics, clinicopathological features, tumor biology, treatment patterns, and drug selections between Eastern and Western gastric cancer patients. Key revisions include a structured template for imaging diagnosis reports, updated standards for molecular marker testing in pathological diagnosis, and an elevated recommendation for neoadjuvant chemotherapy in stage III gastric cancer. For advanced metastatic gastric cancer, the guidelines introduce new recommendations for immunotherapy, anti-angiogenic therapy and targeted drugs, along with updated management strategies for human epidermal growth factor receptor 2 (HER2)-positive and deficient DNA mismatch repair (dMMR)/microsatellite instability-high (MSI-H) patients. Additionally, the guidelines offer detailed screening recommendations for hereditary gastric cancer and an appendix listing drug treatment regimens for various stages of gastric cancer. The 2023 CSCO Clinical Guidelines for Gastric Cancer updates are based on both Chinese and international clinical research and expert consensus to enhance their applicability and relevance in clinical practice, particularly in the heterogeneous healthcare landscape of China, while maintaining a commitment to scientific rigor, impartiality, and timely revisions.</p>\u0000 </section>\u0000 </div>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":"44 1","pages":"127-172"},"PeriodicalIF":16.2,"publicationDate":"2023-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12516","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139063385","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
Development of antibody-drug conjugates in cancer: Overview and prospects 开发癌症抗体药物共轭物:概述与前景
IF 16.2 1区 医学 Q1 ONCOLOGY Pub Date : 2023-12-30 DOI: 10.1002/cac2.12517
Dan-Yun Ruan, Hao-Xiang Wu, Qi Meng, Rui-Hua Xu

In recent years, remarkable breakthroughs have been reported on antibody-drug conjugates (ADCs), with 15 ADCs successfully entering the market over the past decade. This substantial development has positioned ADCs as one of the fastest-growing domains in the realm of anticancer drugs, demonstrating their efficacy in treating a wide array of malignancies. Nonetheless, there is still an unmet clinical need for wider application, better efficacy, and fewer side effects of ADCs. An ADC generally comprises an antibody, a linker and a payload, and the combination has profound effects on drug structure, pharmacokinetic profile and efficacy. Hence, optimization of the key components provides an opportunity to develop ADCs with higher potency and fewer side effects. In this review, we comprehensively reviewed the current development and the prospects of ADC, provided an analysis of marketed ADCs and the ongoing pipelines globally as well as in China, highlighted several ADC platforms and technologies specific to different pharmaceutical enterprises and biotech companies, and also discussed the new related technologies, possibility of next-generation ADCs and the directions of clinical research.

近年来,抗体药物共轭物(ADC)取得了重大突破,在过去十年中,共有 15 种 ADC 成功进入市场。这一重大发展使 ADC 成为抗癌药物领域中发展最快的领域之一,证明了其在治疗各种恶性肿瘤方面的疗效。尽管如此,ADC 在更广泛的应用、更好的疗效和更少的副作用方面仍有未满足的临床需求。ADC 通常由抗体、连接体和有效载荷组成,三者的组合对药物结构、药代动力学特征和疗效有着深远的影响。因此,优化关键成分可为开发药效更高、副作用更小的 ADC 提供机会。在这篇综述中,我们全面回顾了ADC的发展现状和前景,分析了全球和中国已上市的ADC和在研项目,重点介绍了不同制药企业和生物技术公司特有的几种ADC平台和技术,还讨论了相关新技术、下一代ADC的可能性和临床研究方向。
{"title":"Development of antibody-drug conjugates in cancer: Overview and prospects","authors":"Dan-Yun Ruan,&nbsp;Hao-Xiang Wu,&nbsp;Qi Meng,&nbsp;Rui-Hua Xu","doi":"10.1002/cac2.12517","DOIUrl":"10.1002/cac2.12517","url":null,"abstract":"<p>In recent years, remarkable breakthroughs have been reported on antibody-drug conjugates (ADCs), with 15 ADCs successfully entering the market over the past decade. This substantial development has positioned ADCs as one of the fastest-growing domains in the realm of anticancer drugs, demonstrating their efficacy in treating a wide array of malignancies. Nonetheless, there is still an unmet clinical need for wider application, better efficacy, and fewer side effects of ADCs. An ADC generally comprises an antibody, a linker and a payload, and the combination has profound effects on drug structure, pharmacokinetic profile and efficacy. Hence, optimization of the key components provides an opportunity to develop ADCs with higher potency and fewer side effects. In this review, we comprehensively reviewed the current development and the prospects of ADC, provided an analysis of marketed ADCs and the ongoing pipelines globally as well as in China, highlighted several ADC platforms and technologies specific to different pharmaceutical enterprises and biotech companies, and also discussed the new related technologies, possibility of next-generation ADCs and the directions of clinical research.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":"44 1","pages":"3-22"},"PeriodicalIF":16.2,"publicationDate":"2023-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12517","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139063437","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