首页 > 最新文献

MedComm – Oncology最新文献

英文 中文
Targeting LAG-3 restores CD8+ T cell effector function through CD94/NKG2-Qa-1b signaling 靶向 LAG-3 可通过 CD94/NKG2-Qa-1b 信号恢复 CD8+ T 细胞效应功能
Pub Date : 2024-11-24 DOI: 10.1002/mog2.70003
Zhiqiang Wang, Mingzhu Yin, Ge Lou
<p>A recent study published in <i>Cell</i> by Ngiow et al. elucidates the synergistic roles of programmed cell death protein 1 (PD-1) and lymphocyte-activation gene 3 (LAG-3) in CD8<sup>+</sup> T cells.<span><sup>1</sup></span> The authors demonstrate that LAG-3 influences the fate of these cells by sustaining thymocyte selection-associated high mobility group box protein (TOX) expression and modulating the CD94/NKG2-Qa-1b axis in exhausted CD8<sup>+</sup> T cells. This research enhances our understanding of the intricate mechanisms underlying immune checkpoint blockade and paves the way for novel approaches in cancer immunotherapy.</p><p>In chronic viral infections and cancers, CD8<sup>+</sup> T cells, known as the “warriors” of the immune system, frequently experience exhaustion due to persistent antigenic stimulation. This exhaustion results in impaired proliferation and effector function of exhausted CD8<sup>+</sup> T cells (Tex), ultimately leading to immune failure. The defining characteristics of Tex include a distinct TOX-driven transcriptional and epigenetic state, along with the sustained expression of various inhibitory receptors (IRs), such as PD-1, LAG-3, and cytotoxic T-lymphocyte antigen-4 (CTLA-4).<span><sup>2</sup></span> Recent advancements in immune checkpoint blockade therapies that target various IRs have significantly improved tumor treatment outcomes. However, only a subset of tumor patients exhibit sustained clinical responses to monotherapy with PD-1/programmed death-ligand 1 (PD-L1) blockade. Consequently, there is a growing interest in exploring the combination of multiple immune receptors to enhance the efficacy of immunotherapeutic approaches. A recent phase III clinical trial, RELATIVITY-047, demonstrated that combining a LAG-3 antibody (relatlimab) and a PD-1 antibody (nivolumab) exhibits enhanced clinical antitumor effects. However, the precise mechanisms by which PD-1 and LAG-3 regulate Tex function, as well as the interconnections between their signaling pathways, remain unclear.</p><p>Ngiow et al. investigated the effects of LAG-3 and PD-1 on CD8<sup>+</sup> T cells at various stages of chronic infection using the Quad transplantation model. Their findings revealed that CD8<sup>+</sup> T cells lacking PD-1 proliferated significantly faster during the early stages of infection; however, these cells became progressively less sustainable over time. In contrast, cells lacking LAG-3 demonstrated enhanced effector functions, particularly in terms of cytotoxicity and cytokine secretion. This indicates that PD-1 primarily inhibits cell proliferation, while LAG-3 restricts the multifunctionality and killing capacity of effector T cells as they transition into exhausted T cells. The researchers further investigated the mechanism by which LAG-3 influences the differentiation of Tex cells through the regulation of TOX expression. Their findings revealed that LAG-3 deficiency resulted in a significant reduction in TOX expres
Ngiow等人最近在《细胞》(Cell)杂志上发表的一项研究阐明了程序性细胞死亡蛋白1(PD-1)和淋巴细胞活化基因3(LAG-3)在CD8+T细胞中的协同作用1。作者证明,LAG-3通过维持胸腺细胞选择相关的高迁移率组盒蛋白(TOX)表达和调节CD8+T细胞衰竭后的CD94/NKG2-Qa-1b轴来影响这些细胞的命运。在慢性病毒感染和癌症中,CD8+ T 细胞被称为免疫系统的 "战士",它们经常会因持续的抗原刺激而衰竭。这种衰竭导致衰竭的 CD8+ T 细胞(Tex)的增殖和效应功能受损,最终导致免疫失败。Tex 的显著特征包括由 TOX 驱动的转录和表观遗传状态,以及各种抑制受体(IR)的持续表达,如 PD-1、LAG-3 和细胞毒性 T 淋巴细胞抗原-4(CTLA-4)。然而,只有一部分肿瘤患者对 PD-1/程序性死亡配体 1(PD-L1)阻断的单一疗法表现出持续的临床反应。因此,人们越来越有兴趣探索多种免疫受体的联合应用,以提高免疫治疗方法的疗效。最近的一项III期临床试验RELATIVITY-047证明,LAG-3抗体(relatlimab)和PD-1抗体(nivolumab)联合使用可增强临床抗肿瘤效果。然而,PD-1和LAG-3调控Tex功能的确切机制以及它们信号通路之间的相互联系仍不清楚。Ngiow等人利用Quad移植模型研究了LAG-3和PD-1在慢性感染不同阶段对CD8+T细胞的影响。他们的研究结果表明,在感染的早期阶段,缺乏 PD-1 的 CD8+ T 细胞增殖速度明显加快;然而,随着时间的推移,这些细胞的可持续性逐渐减弱。相比之下,缺乏LAG-3的细胞表现出更强的效应功能,尤其是在细胞毒性和细胞因子分泌方面。这表明PD-1主要抑制细胞增殖,而LAG-3在效应T细胞转变为衰竭T细胞时限制了它们的多功能性和杀伤能力。研究人员进一步研究了LAG-3通过调控TOX表达影响Tex细胞分化的机制。他们的研究结果表明,LAG-3的缺乏会导致TOX表达的显著减少,同时伴随着TCF1+ Tex前体细胞的减少。此外,RNA 测序结果表明,LAG-3 缺陷细胞的转录谱更接近效应 T 细胞,显示出细胞毒性相关基因的表达增强。这些基因包括 Klr 家族的多个成员(如 Klrg1 和 Klrd1),表明 LAG-3 在抑制自然杀伤(NK)受体相关基因的表达方面至关重要。生物信息学数据分析进一步显示,LAG-3基因敲除(KO)细胞在与NK细胞介导的细胞毒性相关的通路中表现出富集。这些发现不仅强调了LAG-3在调控Tex细胞功能和分化中的重要作用,还表明LAG-3可能通过调控NK受体影响细胞杀伤能力和免疫监视。研究人员随后利用单细胞 RNA 测序技术研究了 PD-1 KO 和 LAG-3 KO 细胞的转录组特征,并将它们与 TOX 缺失细胞进行了比较。实验结果表明,在LAG-3缺陷细胞中,CD94和Klrd1等与NK受体相关的基因表达上调,这与NK细胞样功能增强有关。这种增强在肿瘤微环境中尤为明显,表明LAG-3调节Tex的一个关键机制是通过抑制CD94/NKG2轴的激活。研究人员观察到,与抑制性受体NK细胞凝集素样受体C亚家族成员1(NKG2A)相比,LAG-3缺陷的Tex细胞表现出更高的活化受体NK细胞2组异构体C/E(NKG2C/E)表达量。这种表达模式的转变使这些细胞能更有效地靶向并消灭微环境中的肿瘤细胞。
{"title":"Targeting LAG-3 restores CD8+ T cell effector function through CD94/NKG2-Qa-1b signaling","authors":"Zhiqiang Wang,&nbsp;Mingzhu Yin,&nbsp;Ge Lou","doi":"10.1002/mog2.70003","DOIUrl":"https://doi.org/10.1002/mog2.70003","url":null,"abstract":"&lt;p&gt;A recent study published in &lt;i&gt;Cell&lt;/i&gt; by Ngiow et al. elucidates the synergistic roles of programmed cell death protein 1 (PD-1) and lymphocyte-activation gene 3 (LAG-3) in CD8&lt;sup&gt;+&lt;/sup&gt; T cells.&lt;span&gt;&lt;sup&gt;1&lt;/sup&gt;&lt;/span&gt; The authors demonstrate that LAG-3 influences the fate of these cells by sustaining thymocyte selection-associated high mobility group box protein (TOX) expression and modulating the CD94/NKG2-Qa-1b axis in exhausted CD8&lt;sup&gt;+&lt;/sup&gt; T cells. This research enhances our understanding of the intricate mechanisms underlying immune checkpoint blockade and paves the way for novel approaches in cancer immunotherapy.&lt;/p&gt;&lt;p&gt;In chronic viral infections and cancers, CD8&lt;sup&gt;+&lt;/sup&gt; T cells, known as the “warriors” of the immune system, frequently experience exhaustion due to persistent antigenic stimulation. This exhaustion results in impaired proliferation and effector function of exhausted CD8&lt;sup&gt;+&lt;/sup&gt; T cells (Tex), ultimately leading to immune failure. The defining characteristics of Tex include a distinct TOX-driven transcriptional and epigenetic state, along with the sustained expression of various inhibitory receptors (IRs), such as PD-1, LAG-3, and cytotoxic T-lymphocyte antigen-4 (CTLA-4).&lt;span&gt;&lt;sup&gt;2&lt;/sup&gt;&lt;/span&gt; Recent advancements in immune checkpoint blockade therapies that target various IRs have significantly improved tumor treatment outcomes. However, only a subset of tumor patients exhibit sustained clinical responses to monotherapy with PD-1/programmed death-ligand 1 (PD-L1) blockade. Consequently, there is a growing interest in exploring the combination of multiple immune receptors to enhance the efficacy of immunotherapeutic approaches. A recent phase III clinical trial, RELATIVITY-047, demonstrated that combining a LAG-3 antibody (relatlimab) and a PD-1 antibody (nivolumab) exhibits enhanced clinical antitumor effects. However, the precise mechanisms by which PD-1 and LAG-3 regulate Tex function, as well as the interconnections between their signaling pathways, remain unclear.&lt;/p&gt;&lt;p&gt;Ngiow et al. investigated the effects of LAG-3 and PD-1 on CD8&lt;sup&gt;+&lt;/sup&gt; T cells at various stages of chronic infection using the Quad transplantation model. Their findings revealed that CD8&lt;sup&gt;+&lt;/sup&gt; T cells lacking PD-1 proliferated significantly faster during the early stages of infection; however, these cells became progressively less sustainable over time. In contrast, cells lacking LAG-3 demonstrated enhanced effector functions, particularly in terms of cytotoxicity and cytokine secretion. This indicates that PD-1 primarily inhibits cell proliferation, while LAG-3 restricts the multifunctionality and killing capacity of effector T cells as they transition into exhausted T cells. The researchers further investigated the mechanism by which LAG-3 influences the differentiation of Tex cells through the regulation of TOX expression. Their findings revealed that LAG-3 deficiency resulted in a significant reduction in TOX expres","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70003","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142707993","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Pharmacokinetics comparison of aumolertinib, osimertinib, gefitinib and their major metabolites in mouse model and NSCLC patients 奥美替尼、奥西莫替尼、吉非替尼及其主要代谢物在小鼠模型和 NSCLC 患者中的药代动力学比较
Pub Date : 2024-11-24 DOI: 10.1002/mog2.70002
Ran Ren, Jiangang Zhang, Yongpeng He, Xianghua Zeng, Yu Zhou, Luyao Shen, Yongsheng Li, Dairong Li, Huakan Zhao

The tyrosine kinase inhibitors (TKIs) of epidermal growth factor receptor (EGFR), such as osimertinib and gefitinib, aumolertinib have been widely used in EGFR mutation-positive non-small cell lung cancer (NSCLC) patients. However, the discrepancy of pharmacokinetic features and distributions in important tissues between these EGFR-TKIs remains obscure. In this study, an ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method with specificity and accuracy was established. After a single equivalent dose ratio or equal dose gavage, aumolertinib displayed the shortest elimination half-life time (t1/2), while it showed the largest area under the concentration–time curve in mouse plasma and bone marrow among these 3 EGFR-TKIs. Furthermore, at the time of reaching maximum concentration (tmax) after single equivalent dose ratio gavage, the concentrations of aumolertinib were significantly higher than that of osimertinib and gefitinib in 9 important tissues of mice. Moreover, after single oral administration, aumolertinib displayed the highest concentration in plasma samples from EGFR mutation-positive NSCLC patients. Collectively, our findings manifest that the bioavailability and tissue distribution features of aumolertinib are superior to those of osimertinib and gefitinib, providing a pharmacokinetic basis for the clinical application of aumolertinib and the development of next-generation EGFR-TKIs.

表皮生长因子受体(EGFR)酪氨酸激酶抑制剂(TKIs),如奥西美替尼和吉非替尼、奥莫乐替尼,已广泛用于EGFR突变阳性的非小细胞肺癌(NSCLC)患者。然而,这些表皮生长因子受体抑制剂(EGFR-TKIs)的药代动力学特征和在重要组织中的分布差异仍不明显。本研究建立了一种具有特异性和准确性的超高效液相色谱-串联质谱(UPLC-MS/MS)方法。在这3种表皮生长因子受体抑制剂中,奥美替尼在小鼠血浆和骨髓中的消除半衰期(t1/2)最短,浓度-时间曲线下面积最大。此外,单次等量比灌胃后达到最大浓度(tmax)时,奥美替尼在小鼠9个重要组织中的浓度明显高于奥希替尼和吉非替尼。此外,单次口服后,表皮生长因子受体突变阳性的 NSCLC 患者血浆样本中的奥美替尼浓度最高。总之,我们的研究结果表明,奥美替尼的生物利用度和组织分布特征优于奥希替尼和吉非替尼,为奥美替尼的临床应用和新一代EGFR-TKIs的开发提供了药代动力学基础。
{"title":"Pharmacokinetics comparison of aumolertinib, osimertinib, gefitinib and their major metabolites in mouse model and NSCLC patients","authors":"Ran Ren,&nbsp;Jiangang Zhang,&nbsp;Yongpeng He,&nbsp;Xianghua Zeng,&nbsp;Yu Zhou,&nbsp;Luyao Shen,&nbsp;Yongsheng Li,&nbsp;Dairong Li,&nbsp;Huakan Zhao","doi":"10.1002/mog2.70002","DOIUrl":"https://doi.org/10.1002/mog2.70002","url":null,"abstract":"<p>The tyrosine kinase inhibitors (TKIs) of epidermal growth factor receptor (EGFR), such as osimertinib and gefitinib, aumolertinib have been widely used in EGFR mutation-positive non-small cell lung cancer (NSCLC) patients. However, the discrepancy of pharmacokinetic features and distributions in important tissues between these EGFR-TKIs remains obscure. In this study, an ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method with specificity and accuracy was established. After a single equivalent dose ratio or equal dose gavage, aumolertinib displayed the shortest elimination half-life time (<i>t</i><sub>1/2</sub>), while it showed the largest area under the concentration–time curve in mouse plasma and bone marrow among these 3 EGFR-TKIs. Furthermore, at the time of reaching maximum concentration (<i>t</i><sub>max</sub>) after single equivalent dose ratio gavage, the concentrations of aumolertinib were significantly higher than that of osimertinib and gefitinib in 9 important tissues of mice. Moreover, after single oral administration, aumolertinib displayed the highest concentration in plasma samples from EGFR mutation-positive NSCLC patients. Collectively, our findings manifest that the bioavailability and tissue distribution features of aumolertinib are superior to those of osimertinib and gefitinib, providing a pharmacokinetic basis for the clinical application of aumolertinib and the development of next-generation EGFR-TKIs.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70002","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142708332","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Epigenetic remodeling under oxidative stress: Mechanisms driving tumor metastasis 氧化应激下的表观遗传重塑:肿瘤转移的驱动机制
Pub Date : 2024-11-14 DOI: 10.1002/mog2.70000
Peilan Peng, Siyuan Qin, Lei Li, Zhijun He, Bowen Li, Edouard C. Nice, Li Zhou, Yunlong Lei

Tumor metastasis is a multistep, inefficient process orchestrated by diverse signaling pathways. Compared to primary tumor cells, disseminated tumor cells inevitably encounter higher oxidative stress in foreign environments. The levels of reactive oxygen species (ROS) fluctuate dynamically during different metastatic stages, adding complexity to the regulation of metastatic progression. Numerous studies suggest that epigenetic remodeling, a key reversible mechanism of gene regulation, plays a critical role in responding to oxidative stress and controlling gene expression profiles that drive metastasis. Despite extensive research, a comprehensive understanding of how oxidative stress impacts metastasis through epigenetic modifications remains elusive, such as DNA methylation, histone modification, ncRNAs, and m6A modification. Epigenetic therapeutic strategies, such as DNMT inhibitors, HDAC inhibitors (HDACis), and miRNA mimics, have shown promise, yet challenges related to immunogenicity, specificity, and delivery also exist. Furthermore, due to limited understanding, some drugs targeting m6A modification have yet to be explored. In this review, we provided an overview of how oxidative stress influences tumor metastatic behavior, summarized the epigenetic mechanisms involved in these processes, and reviewed the latest advancements in epigenetic-targeted therapies, which may pave the way to develop novel strategy for preventing or treating tumor metastasis.

肿瘤转移是一个由多种信号通路协调的多步骤、低效率过程。与原发肿瘤细胞相比,扩散的肿瘤细胞在外来环境中不可避免地会遇到更高的氧化应激。在不同的转移阶段,活性氧(ROS)的水平动态波动,增加了转移进展调控的复杂性。大量研究表明,表观遗传重塑是一种关键的可逆基因调控机制,在应对氧化应激和控制驱动转移的基因表达谱方面发挥着关键作用。尽管进行了广泛的研究,但对氧化应激如何通过表观遗传修饰(如 DNA 甲基化、组蛋白修饰、ncRNA 和 m6A 修饰)影响转移的全面了解仍然遥不可及。表观遗传学治疗策略,如 DNMT 抑制剂、HDAC 抑制剂(HDACis)和 miRNA 模拟物,已显示出良好的前景,但在免疫原性、特异性和递送方面也存在挑战。此外,由于了解有限,一些针对 m6A 修饰的药物还有待探索。在这篇综述中,我们概述了氧化应激如何影响肿瘤转移行为,总结了参与这些过程的表观遗传学机制,并回顾了表观遗传学靶向疗法的最新进展,这些进展可能为开发预防或治疗肿瘤转移的新策略铺平道路。
{"title":"Epigenetic remodeling under oxidative stress: Mechanisms driving tumor metastasis","authors":"Peilan Peng,&nbsp;Siyuan Qin,&nbsp;Lei Li,&nbsp;Zhijun He,&nbsp;Bowen Li,&nbsp;Edouard C. Nice,&nbsp;Li Zhou,&nbsp;Yunlong Lei","doi":"10.1002/mog2.70000","DOIUrl":"https://doi.org/10.1002/mog2.70000","url":null,"abstract":"<p>Tumor metastasis is a multistep, inefficient process orchestrated by diverse signaling pathways. Compared to primary tumor cells, disseminated tumor cells inevitably encounter higher oxidative stress in foreign environments. The levels of reactive oxygen species (ROS) fluctuate dynamically during different metastatic stages, adding complexity to the regulation of metastatic progression. Numerous studies suggest that epigenetic remodeling, a key reversible mechanism of gene regulation, plays a critical role in responding to oxidative stress and controlling gene expression profiles that drive metastasis. Despite extensive research, a comprehensive understanding of how oxidative stress impacts metastasis through epigenetic modifications remains elusive, such as DNA methylation, histone modification, ncRNAs, and m<sup>6</sup>A modification. Epigenetic therapeutic strategies, such as DNMT inhibitors, HDAC inhibitors (HDACis), and miRNA mimics, have shown promise, yet challenges related to immunogenicity, specificity, and delivery also exist. Furthermore, due to limited understanding, some drugs targeting m<sup>6</sup>A modification have yet to be explored. In this review, we provided an overview of how oxidative stress influences tumor metastatic behavior, summarized the epigenetic mechanisms involved in these processes, and reviewed the latest advancements in epigenetic-targeted therapies, which may pave the way to develop novel strategy for preventing or treating tumor metastasis.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70000","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142642294","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Immunotherapy for gastric cancer: Advances and challenges 胃癌免疫疗法:进展与挑战
Pub Date : 2024-11-04 DOI: 10.1002/mog2.92
Pei Zhang, Chenyan Zhang, Xiaoying Li, Chen Chang, Cailing Gan, Tinghong Ye, Dan Cao

Gastric cancer (GC) ranks among the leading causes of cancer-related mortality globally. Often, its initial stages manifest subtly, and the infrequency of routine screenings contributes to late diagnoses in many cases. Systemic treatments for GC include chemotherapy, targeted therapy, and immunotherapy, among which immunotherapy is the first-line standard treatment for advanced GC. In recent years, immunotherapy has seen notable advancements, as evidenced by the Food and Drug Administration's approval of drugs such as nivolumab and pembrolizumab for GC treatment. Additionally, several other drugs are currently under rigorous preclinical and clinical investigation. This review aims to shed light on the recent advancements in immunotherapy for GC, particularly emphasizing the insights gained from phase 2/3 clinical trials that assess the efficacy, safety, and promise of various immunotherapeutic modalities, including immune checkpoint inhibitors, CAR-T-cell therapies, and cancer vaccines, in enhancing patient outcomes. Moreover, this review delves into the intricate immunological framework of GC, focusing on the tumor microenvironment, interactions among immune cells, and the roles of immune checkpoints such as PD-L1. We also address the hurdles and prospective paths forward in the realm of immunotherapy for GC, offering fresh viewpoints on potential therapeutic approaches in this evolving domain.

胃癌(GC)是全球癌症相关死亡的主要原因之一。胃癌的初期症状往往很不明显,而常规筛查又不常见,导致很多病例诊断较晚。GC 的系统治疗包括化疗、靶向治疗和免疫治疗,其中免疫治疗是晚期 GC 的一线标准治疗方法。近年来,免疫疗法取得了显著进展,美国食品和药物管理局已批准 nivolumab 和 pembrolizumab 等药物用于 GC 治疗。此外,目前还有其他几种药物正在接受严格的临床前和临床研究。本综述旨在阐明 GC 免疫疗法的最新进展,特别强调从 2/3 期临床试验中获得的见解,这些临床试验评估了各种免疫治疗模式(包括免疫检查点抑制剂、CAR-T 细胞疗法和癌症疫苗)的疗效、安全性和前景,以提高患者的预后。此外,本综述还深入探讨了 GC 错综复杂的免疫学框架,重点关注肿瘤微环境、免疫细胞之间的相互作用以及 PD-L1 等免疫检查点的作用。我们还探讨了 GC 免疫疗法领域的障碍和前景,为这一不断发展的领域中的潜在治疗方法提供了新的观点。
{"title":"Immunotherapy for gastric cancer: Advances and challenges","authors":"Pei Zhang,&nbsp;Chenyan Zhang,&nbsp;Xiaoying Li,&nbsp;Chen Chang,&nbsp;Cailing Gan,&nbsp;Tinghong Ye,&nbsp;Dan Cao","doi":"10.1002/mog2.92","DOIUrl":"https://doi.org/10.1002/mog2.92","url":null,"abstract":"<p>Gastric cancer (GC) ranks among the leading causes of cancer-related mortality globally. Often, its initial stages manifest subtly, and the infrequency of routine screenings contributes to late diagnoses in many cases. Systemic treatments for GC include chemotherapy, targeted therapy, and immunotherapy, among which immunotherapy is the first-line standard treatment for advanced GC. In recent years, immunotherapy has seen notable advancements, as evidenced by the Food and Drug Administration's approval of drugs such as nivolumab and pembrolizumab for GC treatment. Additionally, several other drugs are currently under rigorous preclinical and clinical investigation. This review aims to shed light on the recent advancements in immunotherapy for GC, particularly emphasizing the insights gained from phase 2/3 clinical trials that assess the efficacy, safety, and promise of various immunotherapeutic modalities, including immune checkpoint inhibitors, CAR-T-cell therapies, and cancer vaccines, in enhancing patient outcomes. Moreover, this review delves into the intricate immunological framework of GC, focusing on the tumor microenvironment, interactions among immune cells, and the roles of immune checkpoints such as PD-L1. We also address the hurdles and prospective paths forward in the realm of immunotherapy for GC, offering fresh viewpoints on potential therapeutic approaches in this evolving domain.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.92","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142587953","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Nanoparticle-encapsuled celastrol-Fe(III): “Single-edged sword” for tumor therapy featuring microenvironment-dependent toxicity 纳米颗粒封装的细胞铁(III):治疗肿瘤的 "单刃剑",具有微环境依赖性毒性
Pub Date : 2024-10-28 DOI: 10.1002/mog2.70001
Yu Liu, Zhiyong Qian
<p>Recently, Li et al. published a study in <i>Science Advances</i> on the use of dual-responsive celastrol (CEL) materials for cancer treatment. The study took advantage of the high adenosine triphosphate (ATP) levels in the tumor microenvironment to modify the interior and surface of CEL, reducing its toxicity to normal tissues and maximizing the efficacy of its cancer treatment which contributed significantly to drug delivery for tumor therapy.<span><sup>1</sup></span></p><p>CEL is a drug with anticancer activity isolated from traditional Chinese medicinal herbs.<span><sup>2</sup></span> It inhibits cancer cell growth and survival signaling by binding to Cdc37 and Hsp90 reaction sites, causing disruption of the Hsp90-Cdc37 binding and folding of some of the client proteins.<span><sup>3</sup></span> However, its water stability, bioavailability, and targeting are undesirable, which limited its clinical application. There are currently two main approaches to promote CEL's therapeutic potential in clinic: 1. synthesizing derivatives by surface modification which mainly focuses on C-20 carboxylic acid functionality, alteration at the A ring, and C-6-modification at the B ring, and 2. adopting nanomedicine delivery systems, where liposomes, polymeric micelles, and nanoparticles (NPs) are commonly used. The second approach, however, requires strictly selected materials. Liposomes, with its ease of aggregation and instability and leakage when packaging drugs, and polymeric micelles, with potential toxicity, unclear drug delivery mechanisms, and poor quality/specification control, demand extra considerations in application. In this study an ATP/ROS dual-responsive CEL derivative was designed for tumor therapy by mainly taking advantage of the environment in which ATP levels inside and outside tumor cells are 10<sup>3</sup> to 10<sup>4</sup> times higher than that around normal tissues.<span><sup>4</sup></span> Surface modification of CEL using metal chelation was performed to address the toxicity to normal tissues, and NPs were selected for packaging to promote the release of CEL when it reaches the tumor site. Compared with liposomes and polymeric micelles, CEL-Fe with ATP/ROS dual-response has a well-defined structural study, low drug toxicity, high stability, and ultimately the ability to respond to the specificity of the tumor microenvironment for effective drug release and treatment which improves the biosafety and biocompatibility of the therapeutic drugs. Fe(III) was chosen to coordinately bond with oxygens on C-2 and C-3 in CEL to produce the less toxic CEL-Fe, which is then coated with polymer membrane consisting of thioketal-containing polyethylene glycol-grafted polymer and polyethylene enamine-modified F127 polymer to elevate its permeability and retention effects.<span><sup>5</sup></span> The high concentration of ATP in tumor tissues disrupts the coordination of Fe(III), restoring the pharmacological toxicity of CEL. In response to high
最近,Li 等人在《科学进展》(Science Advances)杂志上发表了一项关于使用双响应糜蛋白(CEL)材料治疗癌症的研究。该研究利用肿瘤微环境中的高三磷酸腺苷(ATP)水平,对 CEL 的内部和表面进行了改性,降低了其对正常组织的毒性,最大限度地提高了其癌症治疗效果,为肿瘤治疗的药物输送做出了重要贡献。2 它通过与 Cdc37 和 Hsp90 反应位点结合,破坏 Hsp90-Cdc37 的结合和部分客户蛋白的折叠,从而抑制癌细胞的生长和存活信号传导。目前,促进 CEL 临床治疗潜力的方法主要有两种:1. 通过表面改性合成衍生物,主要集中在 C-20 羧酸官能团、A 环的改变和 B 环的 C-6 改性;以及 2. 采用纳米药物传输系统,常用的有脂质体、聚合物胶束和纳米颗粒(NPs)。不过,第二种方法需要严格选择材料。脂质体在包装药物时容易聚集、不稳定和泄漏,而聚合物胶束则有潜在毒性、给药机制不明确、质量/规格控制不佳等问题,因此在应用时需要格外注意。本研究主要利用肿瘤细胞内外的 ATP 水平是正常组织周围的 103-104 倍这一环境,设计了一种 ATP/ROS 双响应 CEL 衍生物用于肿瘤治疗。与脂质体和高分子胶束相比,具有 ATP/ROS 双响应的 CEL-Fe 具有结构研究明确、药物毒性低、稳定性高的特点,最终能够响应肿瘤微环境的特异性,实现有效的药物释放和治疗,提高了治疗药物的生物安全性和生物相容性。选择铁(III)与 CEL 中 C-2 和 C-3 上的氧原子配位结合,生成毒性较低的 CEL-Fe,然后将其包覆在由含硫酮的聚乙二醇接枝聚合物和聚乙烯烯胺改性 F127 聚合物组成的聚合物膜上,以提高其渗透性和保留效果5。肿瘤组织中高浓度的 ATP 破坏了铁(III)的配位,从而恢复了 CEL 的药理毒性。在肿瘤部位高浓度的 ROS 作用下,硫酮基团被降解,促进了药物的释放,从而启动了肿瘤治疗(图 1A):经 CEL、CEL-Fe 和 CEL-Fe-ATP 处理后,对癌细胞和正常细胞的存活率进行了测试。结果表明,CEL-Fe(III)螯合物具有 ATP 依赖性细胞毒性,从而验证了其生物安全性(图 1B、C)。我们通过对 A549 细胞进行 RNA 测序分析,探索了 CEL 和 CEL-Fe 性质转变的机制。PCA 基因表达分析表明,CEL 组和 CEL-Fe+ATP 组聚集在一起,而与 CEL+Fe 组分开。此外,通过计算基因表达量折叠变化和 p 值图,发现 CEL 组和 CEL-Fe+ATP 组的基因表达量无显著差异,而 CEL+Fe 组的基因表达量有差异。这表明铁(III)螯合作用的细胞毒性减弱,而 ATP 作用的细胞毒性恢复。京都基因组百科全书(KEGG)分析和基因组富集分析表明,ER蛋白处理、肿瘤坏死因子信号转导和P53信号转导在CEL和CEL-Fe-ATP处理中富集程度较高,而在CEL-Fe处理中这些通路有很大程度的重叠。这表明CEL的功能与蛋白质的 "生命周期 "有关,这与现有研究中CEL抗肿瘤治疗的机制是一致的。研究人员进一步研究了CEL的毒性机制,并选择Hsp90/Cdc37作为检测模型。分子对接和 MD 模拟显示,CEL 通过占据疏水位点破坏 Hsp90-Cdc37 内部的氢键,导致蛋白质降解,而 CEL-Fe 破坏的氢键数量明显少于 CEL,从而降低了其对蛋白质复合物和细胞的毒性。上述数据为利用这种 CEL 衍生物进行有效的癌症治疗提供了依据(图 1D、E)。组织切片的hema-toxylin 和 eosin(H&amp;E)染色检验了体内生物安全性。 结果表明,CEL 和 CEL NPs 对多个重要组织和器官有明显毒性,而 CEL-Fe 组则没有。这表明金属螯合作用成功地将肿瘤与正常组织器官区分开来。此外,荧光成像显示,在细胞衍生异种移植(CDX)和患者衍生异种移植(PDX)肿瘤模型中,CEL NPs 在肿瘤部位都有很强的存在性,这证明了其优越的生物分散性和在未来临床试验中的可行性。在规定周期后,CEL-Fe NPs 治疗的两种模型小鼠的肿瘤体积最小,肿瘤治疗效果最好。H&amp;E染色和TUNEL检测均显示,CEL-Fe NPs组的肿瘤坏死率名列前茅。外部包装材料在高 ATP/ROS 环境中被 Fenton 反应破坏,暴露出负载的治疗药物 CEL-Fe,CEL-Fe 在高 ATP 浓度的作用下转化为 CEL,毒性得到恢复,在肿瘤部位充分发挥作用。在这项研究中,对 CEL 的改造巧妙地解决了 CEL 在治疗上区分正常组织和癌症组织的问题,实现了在肿瘤部位进行精准治疗的最终目标。这种设计也是目前肿瘤治疗的主要思路之一,即利用肿瘤微环境与正常组织微环境的差异。这种差异包括 ATP 水平、pH 值、氧含量等。希望研究人员能充分利用肿瘤微环境的内在特性,继续研究和开发具有理想疗效、效率和靶向性的药物或给药系统。钱志勇指导。作者声明无利益冲突。
{"title":"Nanoparticle-encapsuled celastrol-Fe(III): “Single-edged sword” for tumor therapy featuring microenvironment-dependent toxicity","authors":"Yu Liu,&nbsp;Zhiyong Qian","doi":"10.1002/mog2.70001","DOIUrl":"https://doi.org/10.1002/mog2.70001","url":null,"abstract":"&lt;p&gt;Recently, Li et al. published a study in &lt;i&gt;Science Advances&lt;/i&gt; on the use of dual-responsive celastrol (CEL) materials for cancer treatment. The study took advantage of the high adenosine triphosphate (ATP) levels in the tumor microenvironment to modify the interior and surface of CEL, reducing its toxicity to normal tissues and maximizing the efficacy of its cancer treatment which contributed significantly to drug delivery for tumor therapy.&lt;span&gt;&lt;sup&gt;1&lt;/sup&gt;&lt;/span&gt;&lt;/p&gt;&lt;p&gt;CEL is a drug with anticancer activity isolated from traditional Chinese medicinal herbs.&lt;span&gt;&lt;sup&gt;2&lt;/sup&gt;&lt;/span&gt; It inhibits cancer cell growth and survival signaling by binding to Cdc37 and Hsp90 reaction sites, causing disruption of the Hsp90-Cdc37 binding and folding of some of the client proteins.&lt;span&gt;&lt;sup&gt;3&lt;/sup&gt;&lt;/span&gt; However, its water stability, bioavailability, and targeting are undesirable, which limited its clinical application. There are currently two main approaches to promote CEL's therapeutic potential in clinic: 1. synthesizing derivatives by surface modification which mainly focuses on C-20 carboxylic acid functionality, alteration at the A ring, and C-6-modification at the B ring, and 2. adopting nanomedicine delivery systems, where liposomes, polymeric micelles, and nanoparticles (NPs) are commonly used. The second approach, however, requires strictly selected materials. Liposomes, with its ease of aggregation and instability and leakage when packaging drugs, and polymeric micelles, with potential toxicity, unclear drug delivery mechanisms, and poor quality/specification control, demand extra considerations in application. In this study an ATP/ROS dual-responsive CEL derivative was designed for tumor therapy by mainly taking advantage of the environment in which ATP levels inside and outside tumor cells are 10&lt;sup&gt;3&lt;/sup&gt; to 10&lt;sup&gt;4&lt;/sup&gt; times higher than that around normal tissues.&lt;span&gt;&lt;sup&gt;4&lt;/sup&gt;&lt;/span&gt; Surface modification of CEL using metal chelation was performed to address the toxicity to normal tissues, and NPs were selected for packaging to promote the release of CEL when it reaches the tumor site. Compared with liposomes and polymeric micelles, CEL-Fe with ATP/ROS dual-response has a well-defined structural study, low drug toxicity, high stability, and ultimately the ability to respond to the specificity of the tumor microenvironment for effective drug release and treatment which improves the biosafety and biocompatibility of the therapeutic drugs. Fe(III) was chosen to coordinately bond with oxygens on C-2 and C-3 in CEL to produce the less toxic CEL-Fe, which is then coated with polymer membrane consisting of thioketal-containing polyethylene glycol-grafted polymer and polyethylene enamine-modified F127 polymer to elevate its permeability and retention effects.&lt;span&gt;&lt;sup&gt;5&lt;/sup&gt;&lt;/span&gt; The high concentration of ATP in tumor tissues disrupts the coordination of Fe(III), restoring the pharmacological toxicity of CEL. In response to high ","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70001","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142541032","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Extracellular vesicles in cancer drug resistance: Mechanistic insights and therapeutic implications 癌症耐药性中的细胞外囊泡:机理认识和治疗意义
Pub Date : 2024-10-24 DOI: 10.1002/mog2.94
Jing Zuo, Han Yan, Siyuan Qin, Yonghao Yan, Shiqi Wang, Li Yang, Jinlin Yang, Jun Yang

Despite significant advancements in the treatment of cancer, therapeutic resistance remains a major hurdle for the achievement of full cures. Besides being typically driven by intratumoral heterogeneity, such acquired resistance also relies heavily on cell-cell communication whereby extracellular vesicles (EVs) carrying resistance-related components can be transferred from drug-resistant cells or nontumorigenic members within tumor microenvironment (TME) to their sensitive neighbors. The cargo and membrane surface proteins of EVs derived from drug-resistant cells and TME cells carry abundant biological information, transferring therapy-resistant capacity to sensitive cancer cells. Hence, a deep understanding of the roles of EVs in cancer therapy resistance will facilitate identifying biomarkers and developing new approaches to restore therapy sensitivity. In this review, we summarize our current understanding regarding the causes and effects of EV-mediated cell–cell communication in drug resistance, with a particular notice on how various kinds of cargoes derived from different cells within TME are linked to the resistant phenotype. We also discuss how this knowledge can contribute to improvements in clinical practice, that is, the opportunities and challenges of EVs in functioning as potential biomarkers in predicting therapeutic resistance and, by extension, EV-based therapy in achieving deeper and longer-lasting clinical responses.

尽管癌症治疗取得了重大进展,但耐药性仍然是实现完全治愈的主要障碍。这种获得性耐药性除了通常由肿瘤内异质性驱动外,还在很大程度上依赖于细胞间的交流,即携带耐药性相关成分的胞外囊泡(EVs)可以从耐药细胞或肿瘤微环境(TME)中的非致瘤细胞转移到敏感的邻近细胞。来自耐药细胞和肿瘤微环境细胞的EVs的载体和膜表面蛋白携带着丰富的生物信息,将耐药能力传递给敏感的癌细胞。因此,深入了解 EVs 在癌症耐药性中的作用将有助于确定生物标志物和开发新的方法来恢复治疗的敏感性。在这篇综述中,我们总结了目前对耐药性中 EV 介导的细胞-细胞通讯的原因和影响的理解,特别关注了源自 TME 内不同细胞的各种载体如何与耐药表型相关联。我们还讨论了这些知识如何有助于改善临床实践,即 EV 作为潜在的生物标记物在预测耐药性方面的机遇和挑战,以及基于 EV 的疗法在实现更深入、更持久的临床反应方面的机遇和挑战。
{"title":"Extracellular vesicles in cancer drug resistance: Mechanistic insights and therapeutic implications","authors":"Jing Zuo,&nbsp;Han Yan,&nbsp;Siyuan Qin,&nbsp;Yonghao Yan,&nbsp;Shiqi Wang,&nbsp;Li Yang,&nbsp;Jinlin Yang,&nbsp;Jun Yang","doi":"10.1002/mog2.94","DOIUrl":"https://doi.org/10.1002/mog2.94","url":null,"abstract":"<p>Despite significant advancements in the treatment of cancer, therapeutic resistance remains a major hurdle for the achievement of full cures. Besides being typically driven by intratumoral heterogeneity, such acquired resistance also relies heavily on cell-cell communication whereby extracellular vesicles (EVs) carrying resistance-related components can be transferred from drug-resistant cells or nontumorigenic members within tumor microenvironment (TME) to their sensitive neighbors. The cargo and membrane surface proteins of EVs derived from drug-resistant cells and TME cells carry abundant biological information, transferring therapy-resistant capacity to sensitive cancer cells. Hence, a deep understanding of the roles of EVs in cancer therapy resistance will facilitate identifying biomarkers and developing new approaches to restore therapy sensitivity. In this review, we summarize our current understanding regarding the causes and effects of EV-mediated cell–cell communication in drug resistance, with a particular notice on how various kinds of cargoes derived from different cells within TME are linked to the resistant phenotype. We also discuss how this knowledge can contribute to improvements in clinical practice, that is, the opportunities and challenges of EVs in functioning as potential biomarkers in predicting therapeutic resistance and, by extension, EV-based therapy in achieving deeper and longer-lasting clinical responses.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.94","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142525118","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
miRNA interplay: Mechanisms and therapeutic interventions in cancer miRNA 相互作用:癌症的机理和治疗干预措施
Pub Date : 2024-10-15 DOI: 10.1002/mog2.93
Zehua Wang, Hangxuan Wang, Shuhan Zhou, Jiasheng Mao, Zhiqing Zhan, Shiwei Duan

MicroRNAs (miRNAs) are key molecules that regulate gene expression. miRNAs regulate protein synthesis by binding to mRNA, influencing processes such as cell proliferation, metastasis, and apoptosis. They play a pivotal role in cancer development. Current research mainly explores miRNA mechanisms and applications, and the techniques underpinning this research are foundational to both basic science and clinical translation. However, no review has comprehensively examined miRNA mechanisms and applications from a technical perspective, creating a need for this work. Advances in RNA sequencing technology, CRISPR/Cas9 technology, and bioinformatics tools have deepened our understanding of miRNA interactions. miRNA can serve as a biomarker for cancer diagnosis and prognosis, with significant clinical potential. The development of miRNA mimics and inhibitors has brought new hope for cancer treatment, especially in reversing cancer drug resistance. This article reviews the vital role of miRNA interactions in cancer occurrence, development, diagnosis, and treatment, providing new perspectives and strategies for personalized medicine and cancer therapy.

微RNA(miRNA)是调控基因表达的关键分子。miRNA通过与mRNA结合调控蛋白质合成,影响细胞增殖、转移和凋亡等过程。它们在癌症的发展过程中起着举足轻重的作用。目前的研究主要探索 miRNA 的机制和应用,这些研究的基础技术对基础科学和临床转化都具有重要意义。然而,目前还没有综述从技术角度对 miRNA 的机制和应用进行全面研究,因此有必要开展这项工作。RNA 测序技术、CRISPR/Cas9 技术和生物信息学工具的进步加深了我们对 miRNA 相互作用的理解。miRNA 模拟物和抑制剂的开发为癌症治疗带来了新希望,尤其是在逆转癌症耐药性方面。本文综述了 miRNA 相互作用在癌症发生、发展、诊断和治疗中的重要作用,为个性化医疗和癌症治疗提供了新的视角和策略。
{"title":"miRNA interplay: Mechanisms and therapeutic interventions in cancer","authors":"Zehua Wang,&nbsp;Hangxuan Wang,&nbsp;Shuhan Zhou,&nbsp;Jiasheng Mao,&nbsp;Zhiqing Zhan,&nbsp;Shiwei Duan","doi":"10.1002/mog2.93","DOIUrl":"https://doi.org/10.1002/mog2.93","url":null,"abstract":"<p>MicroRNAs (miRNAs) are key molecules that regulate gene expression. miRNAs regulate protein synthesis by binding to mRNA, influencing processes such as cell proliferation, metastasis, and apoptosis. They play a pivotal role in cancer development. Current research mainly explores miRNA mechanisms and applications, and the techniques underpinning this research are foundational to both basic science and clinical translation. However, no review has comprehensively examined miRNA mechanisms and applications from a technical perspective, creating a need for this work. Advances in RNA sequencing technology, CRISPR/Cas9 technology, and bioinformatics tools have deepened our understanding of miRNA interactions. miRNA can serve as a biomarker for cancer diagnosis and prognosis, with significant clinical potential. The development of miRNA mimics and inhibitors has brought new hope for cancer treatment, especially in reversing cancer drug resistance. This article reviews the vital role of miRNA interactions in cancer occurrence, development, diagnosis, and treatment, providing new perspectives and strategies for personalized medicine and cancer therapy.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.93","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142439000","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
KMT2C/D deficiency promotes breast cancer metastasis by regulating KDM6A-mediated epigenetic remodeling KMT2C/D 缺乏通过调节 KDM6A 介导的表观遗传重塑促进乳腺癌转移
Pub Date : 2024-10-14 DOI: 10.1002/mog2.95
Zhao Huang, Wei Zhao
<p>On June 26, 2024, Kornelia Polyak's team published an article in Nature Cell Biology that revealed a new mechanism by which histone-lysine N-methyltransferase 2 C (<i>KMT2C)</i> or <i>KMT2D</i> deletion promotes metalloproteinase 3 (MMP3) expression in a lysine-specific demethylase 6 A (KDM6A)-dependent manner, thereby driving brain metastasis (BMs) of triple-negative breast cancer (TNBC). It provides a new perspective for the treatment of TNBC at the epigenetic level.<span><sup>1</sup></span> Breast cancer is the most common neoplasm and the leading cause of tumor-related death in women worldwide. Breast cancer subtypes can be classified according to the expression of estrogen receptors, progesterone receptors, and human epidermal growth factor receptor 2.<span><sup>2</sup></span> TNBC is characterized by a high level of cell invasiveness and visceral metastasis to organs, usually brain, lungs, and liver, with an average survival time of 18 months. Although TNBC accounts for only about 15%–20% of all breast cancers, it is the subtype with the worst prognosis of breast cancer. Before the advent of immunotherapy, systemic chemotherapies including taxanes, anthracyclines and/or platinum were the predominant first-line treatment options for TNBC. However, the median overall survival of metastatic TNBC is only 9–12 months, and the 5-year survival rate is approximately 12%, which is a serious unmet medical need.<span><sup>3</sup></span></p><p>The most commonly mutated gene in BMs of breast cancer has been reported to be <i>KMT2C/D</i>, and the results of gene sequencing showed that <i>KMT2C</i> and <i>KMT2D</i> were significantly reduced in distal metastasis compared to the primary tumor, suggesting that functional loss of <i>KMT2C/D</i> plays a role in BMs of breast cancer. <i>KMT2C</i> and <i>KMT2D</i> catalyze the methylation of unmethylated H3K4 sites to form H3K4me1, which can be enriched in active enhancer and promoter regions. This modification recruits other histone modifying enzymes, including histone H3K27 acetyltransferases (such as P300) and demethylases (such as KDM6A), which are essential for the regulation of gene expression. As the core components of the SET1-associated protein epigenetic regulatory complex (COMPASS), <i>KMT2C</i> and <i>KMT2D</i> profoundly regulate the epigenetic landscape.<span><sup>4</sup></span> However, it is still unclear how <i>KMT2C</i>/<i>D</i> mutations affect the epigenetic and transcriptomic landscape to promote tumorigenesis.</p><p>Recently, Marco Seehawer and colleagues from Dana-Farber Cancer Institute in the United States found that the loss of <i>KMT2C</i> or <i>KMT2D</i> can lead to metastasis (especially BMs) in a nonmetastatic TNBC mouse model. Mechanistic studies revealed that the loss of <i>KMT2C</i> or <i>KMT2D</i> promotes the expression of matrix MMP3 in a KDM6A-dependent manner, thereby driving BMs of TNBC.<span><sup>1</sup></span> This study provides not only a new perspective for the tre
2024年6月26日,Kornelia Polyak团队在《自然-细胞生物学》(Nature Cell Biology)杂志上发表文章,揭示了组蛋白-赖氨酸N-甲基转移酶2 C(KMT2C)或KMT2D缺失以赖氨酸特异性去甲基化酶6 A(KDM6A)依赖方式促进金属蛋白酶3(MMP3)表达,从而驱动三阴性乳腺癌(TNBC)脑转移(BMs)的新机制。1 乳腺癌是最常见的肿瘤,也是全球妇女因肿瘤死亡的主要原因。乳腺癌亚型可根据雌激素受体、孕激素受体和人表皮生长因子受体 2 的表达情况进行分类。2 TNBC 的特点是细胞侵袭性强,可内脏转移至器官,通常是脑、肺和肝,平均生存时间为 18 个月。虽然 TNBC 只占所有乳腺癌的 15%-20%,但却是预后最差的乳腺癌亚型。在免疫疗法出现之前,包括紫杉类、蒽环类和/或铂类在内的全身化疗是 TNBC 的主要一线治疗方案。然而,转移性 TNBC 的中位总生存期仅为 9-12 个月,5 年生存率约为 12%,这是一个严重的未满足的医疗需求。3 有报道称,乳腺癌骨髓瘤中最常见的突变基因是 KMT2C/D,基因测序结果显示,与原发肿瘤相比,KMT2C 和 KMT2D 在远端转移瘤中明显减少,这表明 KMT2C/D 的功能缺失在乳腺癌骨髓瘤中发挥作用。KMT2C 和 KMT2D 可催化未甲基化的 H3K4 位点的甲基化,形成 H3K4me1,这种甲基化可富集在活跃的增强子和启动子区域。这种修饰会招募其他组蛋白修饰酶,包括组蛋白 H3K27 乙酰转移酶(如 P300)和去甲基化酶(如 KDM6A),它们对基因表达的调控至关重要。最近,美国丹娜-法伯癌症研究所的 Marco Seehawer 及其同事发现,在非转移性 TNBC 小鼠模型中,KMT2C 或 KMT2D 的缺失可导致肿瘤转移(尤其是骨髓瘤)。机理研究发现,KMT2C或KMT2D的缺失会以KDM6A依赖的方式促进基质MMP3的表达,从而驱动TNBC的BMs1。这项研究不仅为从表观遗传学水平治疗TNBC提供了新的视角,也为开发针对BMs的靶向治疗策略提供了潜在靶点。为了研究 KMT2C/D 缺失在乳腺肿瘤中的作用,他们将标记有 H2B-mCherry 的非转移性小鼠乳腺肿瘤细胞系 168FARN 注射到小鼠的乳腺脂肪垫(MFP)中,发现虽然正位肿瘤生长不受影响,但注射了 KMT2C 或 KMT2D 基因敲除(KO)168FARN 细胞的小鼠的肺、肝、脑中出现了 mCherry+ 微转移病灶。随后,研究人员对注射了 MFP 的 168FARN 衍生原发肿瘤进行了单细胞 RNA 测序(scRNA-seq)。差异表达基因分析发现,Ly6a、Bst2、Ifi27l2a和Stat1的表达在KO肿瘤细胞中显著上调,这表明干扰素信号的潜在激活产生了一种促炎环境。此外,他们还发现,KMT2C和KMT2D的缺失对肿瘤微环境中不同类型的细胞亚群有特定的影响。为了剖析KMT2C或KMT2D缺失引起的细胞内在变化,Seehawer等人重点研究了作为KMT2C/D直接靶标的组蛋白。利用定量组蛋白质谱、免疫印迹和染色质免疫沉淀测序(ChIP-seq)方法,他们发现在这两种KO细胞中,H3K4me1发生了位点特异性变化,但没有发生全局性变化。相比之下,H3K27me3的变化是全局性的,而不是位点特异性的。KDM6A 是一种 H3K27me3 去甲基化酶,是 COMPASS 复合物的组成成分之一。KDM6A-ChIP-seq结果显示,KO细胞中的峰值显著增加。随后,Seehawer等人进行了RNA-seq分析,发现KMT2C或KMT2D的缺失会诱导上皮-间质杂交转化(EMT)状态,改变EMT平衡,从而促进转移。值得一提的是,即使在没有EMT的情况下,也能观察到KMT2C/D缺失在促进转移方面的作用。
{"title":"KMT2C/D deficiency promotes breast cancer metastasis by regulating KDM6A-mediated epigenetic remodeling","authors":"Zhao Huang,&nbsp;Wei Zhao","doi":"10.1002/mog2.95","DOIUrl":"https://doi.org/10.1002/mog2.95","url":null,"abstract":"&lt;p&gt;On June 26, 2024, Kornelia Polyak's team published an article in Nature Cell Biology that revealed a new mechanism by which histone-lysine N-methyltransferase 2 C (&lt;i&gt;KMT2C)&lt;/i&gt; or &lt;i&gt;KMT2D&lt;/i&gt; deletion promotes metalloproteinase 3 (MMP3) expression in a lysine-specific demethylase 6 A (KDM6A)-dependent manner, thereby driving brain metastasis (BMs) of triple-negative breast cancer (TNBC). It provides a new perspective for the treatment of TNBC at the epigenetic level.&lt;span&gt;&lt;sup&gt;1&lt;/sup&gt;&lt;/span&gt; Breast cancer is the most common neoplasm and the leading cause of tumor-related death in women worldwide. Breast cancer subtypes can be classified according to the expression of estrogen receptors, progesterone receptors, and human epidermal growth factor receptor 2.&lt;span&gt;&lt;sup&gt;2&lt;/sup&gt;&lt;/span&gt; TNBC is characterized by a high level of cell invasiveness and visceral metastasis to organs, usually brain, lungs, and liver, with an average survival time of 18 months. Although TNBC accounts for only about 15%–20% of all breast cancers, it is the subtype with the worst prognosis of breast cancer. Before the advent of immunotherapy, systemic chemotherapies including taxanes, anthracyclines and/or platinum were the predominant first-line treatment options for TNBC. However, the median overall survival of metastatic TNBC is only 9–12 months, and the 5-year survival rate is approximately 12%, which is a serious unmet medical need.&lt;span&gt;&lt;sup&gt;3&lt;/sup&gt;&lt;/span&gt;&lt;/p&gt;&lt;p&gt;The most commonly mutated gene in BMs of breast cancer has been reported to be &lt;i&gt;KMT2C/D&lt;/i&gt;, and the results of gene sequencing showed that &lt;i&gt;KMT2C&lt;/i&gt; and &lt;i&gt;KMT2D&lt;/i&gt; were significantly reduced in distal metastasis compared to the primary tumor, suggesting that functional loss of &lt;i&gt;KMT2C/D&lt;/i&gt; plays a role in BMs of breast cancer. &lt;i&gt;KMT2C&lt;/i&gt; and &lt;i&gt;KMT2D&lt;/i&gt; catalyze the methylation of unmethylated H3K4 sites to form H3K4me1, which can be enriched in active enhancer and promoter regions. This modification recruits other histone modifying enzymes, including histone H3K27 acetyltransferases (such as P300) and demethylases (such as KDM6A), which are essential for the regulation of gene expression. As the core components of the SET1-associated protein epigenetic regulatory complex (COMPASS), &lt;i&gt;KMT2C&lt;/i&gt; and &lt;i&gt;KMT2D&lt;/i&gt; profoundly regulate the epigenetic landscape.&lt;span&gt;&lt;sup&gt;4&lt;/sup&gt;&lt;/span&gt; However, it is still unclear how &lt;i&gt;KMT2C&lt;/i&gt;/&lt;i&gt;D&lt;/i&gt; mutations affect the epigenetic and transcriptomic landscape to promote tumorigenesis.&lt;/p&gt;&lt;p&gt;Recently, Marco Seehawer and colleagues from Dana-Farber Cancer Institute in the United States found that the loss of &lt;i&gt;KMT2C&lt;/i&gt; or &lt;i&gt;KMT2D&lt;/i&gt; can lead to metastasis (especially BMs) in a nonmetastatic TNBC mouse model. Mechanistic studies revealed that the loss of &lt;i&gt;KMT2C&lt;/i&gt; or &lt;i&gt;KMT2D&lt;/i&gt; promotes the expression of matrix MMP3 in a KDM6A-dependent manner, thereby driving BMs of TNBC.&lt;span&gt;&lt;sup&gt;1&lt;/sup&gt;&lt;/span&gt; This study provides not only a new perspective for the tre","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.95","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142438969","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","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 application of the combination between artificial intelligence and endoscopy in gastrointestinal tumors 人工智能与内窥镜相结合在消化道肿瘤中的应用
Pub Date : 2024-10-04 DOI: 10.1002/mog2.91
Shen Li, Maosen Xu, Yuanling Meng, Haozhen Sun, Tao Zhang, Hanle Yang, Yueyi Li, Xuelei Ma

Gastrointestinal (GI) tumors have always been a major type of malignant tumor and a leading cause of tumor-related deaths worldwide. The main principles of modern medicine for GI tumors are early prevention, early diagnosis, and early treatment, with early diagnosis being the most effective measure. Endoscopy, due to its ability to visualize lesions, has been one of the primary modalities for screening, diagnosing, and treating GI tumors. However, a qualified endoscopist often requires long training and extensive experience, which to some extent limits the wider use of endoscopy. With advances in data science, artificial intelligence (AI) has brought a new development direction for the endoscopy of GI tumors. AI can quickly process large quantities of data and images and improve diagnostic accuracy with some training, greatly reducing the workload of endoscopists and assisting them in early diagnosis. Therefore, this review focuses on the combined application of endoscopy and AI in GI tumors in recent years, describing the latest research progress on the main types of tumors and their performance in clinical trials, the application of multimodal AI in endoscopy, the development of endoscopy, and the potential applications of AI within it, with the aim of providing a reference for subsequent research.

胃肠道(GI)肿瘤一直是恶性肿瘤的主要类型,也是全球肿瘤相关死亡的主要原因。现代医学治疗消化道肿瘤的主要原则是早期预防、早期诊断和早期治疗,其中早期诊断是最有效的措施。内镜检查由于能够直观地观察病灶,一直是筛查、诊断和治疗消化道肿瘤的主要方式之一。然而,一名合格的内镜医师往往需要长期的培训和丰富的经验,这在一定程度上限制了内镜的广泛应用。随着数据科学的发展,人工智能(AI)为消化道肿瘤内镜检查带来了新的发展方向。人工智能可以快速处理大量数据和图像,并通过一定的训练提高诊断准确率,大大减轻内镜医师的工作量,协助他们进行早期诊断。因此,本综述重点关注近年来内镜与人工智能在消化道肿瘤中的结合应用,介绍了主要肿瘤类型及其临床表现、多模态人工智能在内镜中的应用、内镜的发展以及人工智能在其中的潜在应用等方面的最新研究进展,旨在为后续研究提供参考。
{"title":"The application of the combination between artificial intelligence and endoscopy in gastrointestinal tumors","authors":"Shen Li,&nbsp;Maosen Xu,&nbsp;Yuanling Meng,&nbsp;Haozhen Sun,&nbsp;Tao Zhang,&nbsp;Hanle Yang,&nbsp;Yueyi Li,&nbsp;Xuelei Ma","doi":"10.1002/mog2.91","DOIUrl":"https://doi.org/10.1002/mog2.91","url":null,"abstract":"<p>Gastrointestinal (GI) tumors have always been a major type of malignant tumor and a leading cause of tumor-related deaths worldwide. The main principles of modern medicine for GI tumors are early prevention, early diagnosis, and early treatment, with early diagnosis being the most effective measure. Endoscopy, due to its ability to visualize lesions, has been one of the primary modalities for screening, diagnosing, and treating GI tumors. However, a qualified endoscopist often requires long training and extensive experience, which to some extent limits the wider use of endoscopy. With advances in data science, artificial intelligence (AI) has brought a new development direction for the endoscopy of GI tumors. AI can quickly process large quantities of data and images and improve diagnostic accuracy with some training, greatly reducing the workload of endoscopists and assisting them in early diagnosis. Therefore, this review focuses on the combined application of endoscopy and AI in GI tumors in recent years, describing the latest research progress on the main types of tumors and their performance in clinical trials, the application of multimodal AI in endoscopy, the development of endoscopy, and the potential applications of AI within it, with the aim of providing a reference for subsequent research.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.91","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142404314","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Extensive infiltration of CD8+ T cells and M1 macrophages is beneficial for multiple cancer patients undergoing chemotherapy CD8+ T 细胞和 M1 巨噬细胞的广泛浸润对接受化疗的多发性癌症患者有益
Pub Date : 2024-09-20 DOI: 10.1002/mog2.89
Yuquan Bai, He Xu, Minzhang Guo, Liang Xia, Senyi Deng

Not all patients can benefit from chemotherapy due to the various of tumor type, stage, location, and the different distribution of immune cells in tumor immune microenvironment (TIME). Immune cells are widely involved in every step of cancer progression, including immune escape, metastasis, drug response, and prognosis. In this study, we explored the transcriptome data of 10 solid tumors treated with chemotherapy to identify the role of immune cells. We downloaded the transcriptome and mutation data of 10 cancers from TCGA databases, and used ESTIMATE and CIBERSORT algorithms to assess the proportion of immune cells in the TIME. According to the proportion of specific immune cell infiltration (SICI) of CD8+ T cells and M1 macrophages to group the patients, we found that compared with the SICI low and medium groups, the SICI high group had a larger tumor mutation burden, more gene mutations with targeted drugs, more activation of immune checkpoints (PD-1, PD-L1, CTLA-4, LAG-3, TIM-3, and TIGIT), and immune molecules (CD8a, CD80, CD86, TLR2, HLA-A, HLA-B, and CD11a) (p < 0.05). Therefore, we can select an appropriate treatment for patients by clarifying the proportion of immune infiltration of CD8+ T cells and M1 macrophages in the TIME.

由于肿瘤类型、分期、位置以及免疫细胞在肿瘤免疫微环境(TIME)中的分布不同,并非所有患者都能从化疗中获益。免疫细胞广泛参与癌症进展的每一步,包括免疫逃逸、转移、药物反应和预后。在这项研究中,我们探索了 10 例接受化疗的实体瘤的转录组数据,以确定免疫细胞的作用。我们从TCGA数据库中下载了10种癌症的转录组和突变数据,并使用ESTIMATE和CIBERSORT算法评估了免疫细胞在TIME中的比例。根据CD8+ T细胞和M1巨噬细胞的特异性免疫细胞浸润(SICI)比例对患者进行分组,我们发现,与SICI低组和中组相比,SICI高组的肿瘤突变负荷更大、免疫检查点(PD-1、PD-L1、CTLA-4、LAG-3、TIM-3 和 TIGIT)和免疫分子(CD8a、CD80、CD86、TLR2、HLA-A、HLA-B 和 CD11a)的活化程度更高(P <; 0.05).因此,我们可以通过明确 TIME 中 CD8+ T 细胞和 M1 巨噬细胞的免疫浸润比例来为患者选择合适的治疗方法。
{"title":"Extensive infiltration of CD8+ T cells and M1 macrophages is beneficial for multiple cancer patients undergoing chemotherapy","authors":"Yuquan Bai,&nbsp;He Xu,&nbsp;Minzhang Guo,&nbsp;Liang Xia,&nbsp;Senyi Deng","doi":"10.1002/mog2.89","DOIUrl":"https://doi.org/10.1002/mog2.89","url":null,"abstract":"<p>Not all patients can benefit from chemotherapy due to the various of tumor type, stage, location, and the different distribution of immune cells in tumor immune microenvironment (TIME). Immune cells are widely involved in every step of cancer progression, including immune escape, metastasis, drug response, and prognosis. In this study, we explored the transcriptome data of 10 solid tumors treated with chemotherapy to identify the role of immune cells. We downloaded the transcriptome and mutation data of 10 cancers from TCGA databases, and used ESTIMATE and CIBERSORT algorithms to assess the proportion of immune cells in the TIME. According to the proportion of specific immune cell infiltration (SICI) of CD8<sup>+</sup> T cells and M1 macrophages to group the patients, we found that compared with the SICI low and medium groups, the SICI high group had a larger tumor mutation burden, more gene mutations with targeted drugs, more activation of immune checkpoints (PD-1, PD-L1, CTLA-4, LAG-3, TIM-3, and TIGIT), and immune molecules (CD8a, CD80, CD86, TLR2, HLA-A, HLA-B, and CD11a) (<i>p</i> &lt; 0.05). Therefore, we can select an appropriate treatment for patients by clarifying the proportion of immune infiltration of CD8<sup>+</sup> T cells and M1 macrophages in the TIME.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 3","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.89","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142273242","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","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
MedComm – Oncology
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