首页 > 最新文献

MedComm – Oncology最新文献

英文 中文
Targeting iron–sulfur cluster assembly scaffold protein as a vulnerability in KRAS-activated pancreatic ductal adenocarcinoma 靶向铁硫簇组装支架蛋白是 KRAS 激活的胰腺导管腺癌的一个薄弱环节
Pub Date : 2024-01-29 DOI: 10.1002/mog2.65
Huarui Cai, Hongjuan Cui, Erhu Zhao

In two recent publications in Science1 and Cell Discovery,2 researchers have discovered that iron–sulfur cluster assembly scaffold protein (ISCU) plays crucial roles on maintaining glutathione (GSH) homeostasis, α-KG catabolism, DNA methylation and so on. These groundbreaking findings not only establish crucial connections among iron–sulfur (Fe–S) metabolism, GSH homeostasis, alpha-ketoglutarate (α-KG) catabolism, DNA methylation, and tumor growth but also emphasize the significant and integrated regulation of mitochondrial function and gene expression by ISCU (Figure 1).

Liu et al.1 unveiled the vital role of ISCU in maintaining optimal mitochondrial functions by restoring the balance between iron and GSH. In addition, a previous study3 has provided evidence that glutamine, a precursor of GSH, promotes tumor growth through a KRAS-regulated metabolic pathway in pancreatic ductal adenocarcinoma (PDAC), thereby suggesting that ISCU potentially influences tumor development by modulating GSH homeostasis. Ren et al.2 uncovered a previously undisclosed association between Fe–S metabolism and tumor growth in kirsten rat sarcoma viral oncogene homolog (KRAS)-activated PDAC. Such findings offer a promising avenue for the treatment of KRAS-activated PDAC, as well as for interventions targeting ISCU-mediated cellular processes.

PDAC is a highly lethal disease with a mounting incidence and unfavorable prognostic outcomes, underscoring the urgent requirement for the development of efficacious therapeutic approaches. Thus, accurate biomarkers to help stratify risk would greatly improve current diagnostic and decision-making dilemmas. Fe–S clusters are ancient, ubiquitous metal cofactors that possess various physiological functions in antioxidant, iron regulation, the tricarboxylic acid (TCA) cycle, and many other metabolic reactions. Accumulated evidence suggests that abnormal Fe–S clusters assembly pathways lead to mitochondrial dysfunction and cause various diseases, particularly with an impact on tumor development.4 For instance, nitrogen fixation gene 1 (NFS1) cysteine desulfurase, as an indispensable protein in Fe–S cluster biogenesis, plays a vital role in mitochondrial metabolic reprogramming. In human colorectal cancer, NFS1 can suppress PANoptosis under oxaliplatin chemotherapy, and NFS1 high expression was linked to unfavorable survival outcomes and hyposensitivity to chemotherapy in patients.5 In addition, succinate dehydrogenase complex iron–sulfur subunit B (SDHB) is also an Fe–S cluster protein consisting of three Fe–S clusters. In SDHB-deficient cancer cells, succinate levels are elevated, resulting in hypermethylation of histones and DNA, and glutamine becomes the primary source of TCA cycle metabolites via reductive carboxylation.6

No

此外,Ren 等人2 进一步证实,ISCU 的缺失抑制了 PDAC 细胞中所有 Fe-S 依赖性线粒体蛋白的表达,包括二氢脂酰胺 S-琥珀酰基转移酶(DLST)和乌头酶 2(ACO2)。值得注意的是,KRAS 只改变了随脂酰化而稳定的 DLST 蛋白水平,而没有改变其信使 RNA 水平。随后的研究表明,KRAS 缺失导致的 DLST 蛋白水平和 ACO2 活性的降低可被 ISCU 的过表达所抵消。虽然 KRAS 沉默导致的 ISCU 减少不足以抑制 ACO2 蛋白水平,但却足以导致 ACO2 活性的降低,这表明由 ISCU 控制的 ACO2 的稳定性只有在特定的 Fe-S 簇阈值下才会降低。作为一种初级代谢产物,α-KG 具有多种功能,如作为二氧化酶的重要辅助因子:10-11 转化甲基胞嘧啶二氧化酶 1、2、3(TET1、TET2、TET3),可催化 5mC 氧化并调节 DNA 甲基化。Ren等人2发现,活化的KRAS通过控制ISCU促进的α-KG分解和TET3抑制作用增强了DNA甲基化。8 因此,ISCU 介导的 DNA 5mC 水平升高可能会改变许多基因的表达。Ren 等人2 通过全基因组亚硫酸氢盐测序发现,ISCU 沉默改变了 103 个差异表达基因。其中值得注意的是,DNA 聚合酶 I(POLA1)具有在细胞周期 S 期启动 DNA 合成的能力。最后,他们证实,5mC甲基化失调会导致转录异常(如POLA1的激活),这可能会导致PDAC的致癌转化。此外,通过添加α-KG竞争性代谢抑制剂和创造缺氧条件以缓解电子传递链失调,Ren等人2发现,ISCU以α-KG依赖的方式控制细胞增殖。某些临床药物,如用于治疗骨髓增生异常综合征的地西他滨,也是 DNA 甲基转移酶(DNMT)抑制剂。9 Ren 等人 2 发现,ISCU 低表达细胞对地西他滨不敏感,这表明 ISCU 的表达水平有可能成为使用 DNMT 抑制剂治疗 PDAC 的分子生物标志物。总之,Liu 等人1 和 Ren 等人2 提供了令人信服的证据,证明 ISCU 在维持 GSH 稳态、α-KG 分解和 DNA 甲基化等方面发挥着关键作用。此外,作者还发现了PDAC中Fe-S代谢与肿瘤生长之间有趣的联系,为PDAC的发病机制,尤其是KRAS激活突变癌症亚型的发病机制提供了新的见解。值得注意的是,在 KRAS 激活的 PDAC 中,ISCU 被鉴定为 DNA 5mC 水平的调节因子。这些发现不仅为现有DNMT抑制剂的潜在再利用提出了建议,也为新型抑制剂的开发打开了大门,为PDAC的临床干预提供了一条充满希望的途径:概念化(等同);可视化(主要);写作-原稿(主要)。崔红娟指导(等同);撰写-审阅和编辑(支持)。赵二虎构思(等同);经费获取(牵头);指导(等同);撰写-审阅和编辑(牵头)。最终稿件已由所有作者批准。作者声明无利益冲突。
{"title":"Targeting iron–sulfur cluster assembly scaffold protein as a vulnerability in KRAS-activated pancreatic ductal adenocarcinoma","authors":"Huarui Cai,&nbsp;Hongjuan Cui,&nbsp;Erhu Zhao","doi":"10.1002/mog2.65","DOIUrl":"https://doi.org/10.1002/mog2.65","url":null,"abstract":"<p>In two recent publications in Science<span><sup>1</sup></span> and Cell Discovery,<span><sup>2</sup></span> researchers have discovered that iron–sulfur cluster assembly scaffold protein (ISCU) plays crucial roles on maintaining glutathione (GSH) homeostasis, α-KG catabolism, DNA methylation and so on. These groundbreaking findings not only establish crucial connections among iron–sulfur (Fe–S) metabolism, GSH homeostasis, alpha-ketoglutarate (α-KG) catabolism, DNA methylation, and tumor growth but also emphasize the significant and integrated regulation of mitochondrial function and gene expression by ISCU (Figure 1).</p><p>Liu et al.<span><sup>1</sup></span> unveiled the vital role of ISCU in maintaining optimal mitochondrial functions by restoring the balance between iron and GSH. In addition, a previous study<span><sup>3</sup></span> has provided evidence that glutamine, a precursor of GSH, promotes tumor growth through a KRAS-regulated metabolic pathway in pancreatic ductal adenocarcinoma (PDAC), thereby suggesting that ISCU potentially influences tumor development by modulating GSH homeostasis. Ren et al.<span><sup>2</sup></span> uncovered a previously undisclosed association between Fe–S metabolism and tumor growth in kirsten rat sarcoma viral oncogene homolog (KRAS)-activated PDAC. Such findings offer a promising avenue for the treatment of KRAS-activated PDAC, as well as for interventions targeting ISCU-mediated cellular processes.</p><p>PDAC is a highly lethal disease with a mounting incidence and unfavorable prognostic outcomes, underscoring the urgent requirement for the development of efficacious therapeutic approaches. Thus, accurate biomarkers to help stratify risk would greatly improve current diagnostic and decision-making dilemmas. Fe–S clusters are ancient, ubiquitous metal cofactors that possess various physiological functions in antioxidant, iron regulation, the tricarboxylic acid (TCA) cycle, and many other metabolic reactions. Accumulated evidence suggests that abnormal Fe–S clusters assembly pathways lead to mitochondrial dysfunction and cause various diseases, particularly with an impact on tumor development.<span><sup>4</sup></span> For instance, nitrogen fixation gene 1 (NFS1) cysteine desulfurase, as an indispensable protein in Fe–S cluster biogenesis, plays a vital role in mitochondrial metabolic reprogramming. In human colorectal cancer, NFS1 can suppress PANoptosis under oxaliplatin chemotherapy, and NFS1 high expression was linked to unfavorable survival outcomes and hyposensitivity to chemotherapy in patients.<span><sup>5</sup></span> In addition, succinate dehydrogenase complex iron–sulfur subunit B (SDHB) is also an Fe–S cluster protein consisting of three Fe–S clusters. In SDHB-deficient cancer cells, succinate levels are elevated, resulting in hypermethylation of histones and DNA, and glutamine becomes the primary source of TCA cycle metabolites via reductive carboxylation.<span><sup>6</sup></span></p><p>No","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.65","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139655410","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
Lysosome-targeting chimera (LYTAC): A silver bullet for targeted degradation of oncogenic membrane proteins 溶酶体靶向嵌合体(LYTAC):靶向降解致癌膜蛋白的银弹
Pub Date : 2024-01-14 DOI: 10.1002/mog2.64
Qingquan Zheng, Jiawei Guo, Rui Ma, Wenchen Pu

Recently, the group of Prof. Carolyn Bertozzi, a laureate of the Nobel Prize in chemistry 2022, reported the detailed mechanism of lysosome-targeting chimera (LYTAC) in the journal of Science,1 after the publication of their first LYTAC molecule in Nature in 2020.2 The establishment of LYTAC, a subtype of targeted protein degradation technology, expands the scope of protein degradation to extracellular and membrane-associated targets, and Bertozzi group's new discovery is expected to accelerate the development of LYTAC in cancer therapy.

Cell membranes play a critical role in various cellular processes, including signaling transduction, cell adhesion, transport of biomolecules and immunity. Proteins embedded in or associated with the cell membrane are key executants of the function of cell membrane, and their dysregulation contributes to tumorigenesis and development of human cancers.3 For example, epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase for epithelial growth factor (EGF) and transforming growth factor α (TGF-α), belonging to the ErbB receptor family. Activation of EGFR signaling promotes cell proliferation, survival, angiogenesis and metastasis of diverse malignancies.3 Moreover, hepatocyte growth factor receptor (c-Met, HGFR) is another oncogenic receptor tyrosine kinase in diverse cancers. Upon the binding to hepatocyte growth factor (HGF), c-Met is activated via autophosphorylation, leading to the initiation of oncogenic downstream signaling cascades, such as PI3K/AKT and RAS/ERK pathways.3 Given their central role in cancer-promoting processes, EGFR and c-Met has become attractive targets for cancer therapies. Small-molecule tyrosine kinase inhibitors (EGFR: gefitinib, afatinib, osimertinib, etc.; c-Met: capmatinib, tepotinib, savolitinib, etc.) and monoclonal antibodies (EGFR: cetuximab, panitumumab; EGFR/c-Met: amivantamab; c-Met: emibetuzumab), have been developed and approved for the treatment of various cancers, including lung and colorectal cancers (Figure 1A). But severe acquired resistance (e.g., via EGFR mutations) and limited therapeutic efficacy (slightly prolonged overall survival) of these treatments restrict their clinical benefit for patients. Moreover, nonenzymatic function of membrane proteins, such as protein–protein interactions, could not be interfered with by kinase inhibitors or monoclonal antibodies, calling for new strategies to control these oncogenic membrane proteins.

Targeted protein degradation (TPD) is a therapeutic approach that aims to selectively remove disease-causing or undesirable proteins from cells by inducing their degradation, with multiple therapies entering clinical trials and targeting proteins that are previously considered “undruggable.”4 There are two main protein degradation mechanisms within cel

最近,继 2020 年在《自然》(Nature)杂志上发表首个 LYTAC 分子之后,2022 年诺贝尔化学奖得主卡罗琳-贝托齐(Carolyn Bertozzi)教授的研究小组又在《科学》(Science)杂志1 上报告了溶酶体靶向嵌合体(LYTAC)的详细机制。2 LYTAC 是靶向蛋白质降解技术的一个亚型,它的建立将蛋白质降解的范围扩大到细胞外和与膜相关的靶点,Bertozzi 小组的新发现有望加速 LYTAC 在癌症治疗中的发展。细胞膜在信号转导、细胞粘附、生物大分子运输和免疫等各种细胞过程中发挥着关键作用。3 例如,表皮生长因子受体(EGFR)是上皮生长因子(EGF)和转化生长因子α(TGF-α)的受体酪氨酸激酶,属于 ErbB 受体家族。表皮生长因子受体信号激活可促进多种恶性肿瘤的细胞增殖、存活、血管生成和转移。3 此外,肝细胞生长因子受体(c-Met,HGFR)也是多种癌症中的另一种致癌受体酪氨酸激酶。与肝细胞生长因子(HGF)结合后,c-Met 通过自身磷酸化被激活,导致致癌下游信号级联的启动,如 PI3K/AKT 和 RAS/ERK 通路。小分子酪氨酸激酶抑制剂(表皮生长因子受体:吉非替尼、阿法替尼、奥西莫替尼等;c-Met:卡帕替尼、替泊替尼、沙夫利替尼等)和单克隆抗体(表皮生长因子受体:西妥昔单抗、帕尼妥单抗;表皮生长因子受体/c-Met:阿米万坦单抗;c-Met:埃米特珠单抗)已被开发并批准用于治疗各种癌症,包括肺癌和结直肠癌(图 1A)。但是,这些疗法严重的获得性耐药性(如通过表皮生长因子受体突变)和有限的疗效(略微延长总生存期)限制了它们对患者的临床益处。此外,激酶抑制剂或单克隆抗体无法干扰膜蛋白的非酶功能,如蛋白质与蛋白质之间的相互作用,这就要求采用新策略来控制这些致癌膜蛋白。靶向蛋白降解(TPD)是一种治疗方法,旨在通过诱导细胞内致病蛋白或不良蛋白的降解,有选择性地将其从细胞中清除,目前已有多种疗法进入临床试验阶段,其靶向蛋白都是以前被认为 "不可药用 "的蛋白。通过异功能分子触发化学诱导接近形成三元复合物组装,目前已开发出各种基于蛋白酶体(PROTAC、分子胶等)、溶酶体(LYTAC、AUTAC、ATTEC 等)或两者(PROTAB)的 TPD 技术。例如,Jang 等人开发了一种异位表皮生长因子受体蛋白水解靶向嵌合体(PROTAC)--DDC-01-163,该嵌合体对各种临床相关的表皮生长因子受体突变体(L858R/T790M)具有选择性活性,可作为单药或与 ATP 位点抑制剂奥希替尼联合使用(图 1B 左)。5 最近,Marei 等人报道了蛋白水解靶向抗体(PROTABs,一类双特异性抗体),该抗体将细胞表面 E3 泛素连接酶(RNF43 或 ZNRF3)与跨膜蛋白(IGF1R、HER2 和 PD-L1)拴在一起,通过 UPS 和溶酶体途径进行靶向降解,为快速开发强效、生物可用且具有组织选择性的膜蛋白降解剂提供了一种策略。2020 年,Bertozzi 小组制备了双功能分子,其中包括与化学合成的糖肽配体共轭的抗体,糖肽配体是不依赖阳离子的 6-磷酸甘露糖受体(CI-M6PR,一种细胞表面溶酶体关闭受体)的激动剂。这些共轭物能识别 CI-M6PR 和靶蛋白的胞外结构域,诱导溶酶体介导的膜蛋白靶向降解,如表皮生长因子受体、CD71、程序性死亡配体 1 (PD-L1) 和脂蛋白 E4。值得注意的是,CRISPR 干扰筛选的数据表明,在细胞系中,CI-M6PR 介导的货物内化参与其中,并发现外囊复合体参与了这些过程。因此,这些证据证明了通过激活化学诱导的接近性,通过溶酶体降解膜蛋白的可行性。因此,这种技术被称为溶酶体靶向嵌合体(LYTACs)。 2 然而,调控 LYTACs 劫持溶酶体机制降解膜蛋白行为的细胞特征在很大程度上尚属未知。为此,Bertozzi 小组随后采用无偏见的全基因组 CRISPR 基因敲除筛选方法,并辅以蛋白质组学研究,绘制了人体细胞中 LYTAC 介导的膜蛋白降解的关键调控因子图。结果表明,抑制 retromer 基因(如 VPS35、SNX3、VPS29 和 VPS26A)以减少 LYTAC 的循环,可增强目标降解。此外,参与cullin3 (CUL3)内切酶化的基因,如CUL3、UBA3和CAND1,促进了E3连接酶的活性以及LYTAC-靶蛋白复合物向溶酶体的转运。因此,Neddylated CUL3 的水平可作为 LYTAC 疗效的预测指标。此外,膜CI-M6PR受体部分被内源性6-磷酸甘露糖(M6P)修饰的溶酶体糖蛋白啮合。阻断 M6P 生物合成基因(如 ALGO12、GNPTAB)可提高未被占用受体的比例,增加 LYTAC 受体的内化和细胞表面蛋白(包括表皮生长因子受体和 c-Met)的降解。随着基于噬菌体展示、杂交瘤细胞和单 B 细胞技术的单克隆抗体技术的发展和成熟,特异性单克隆抗体的发现和优化变得非常高效。此外,LYTAC 的合成和表征也可以遵循抗体药物共轭物(ADC)的路线,因为它们具有相似的化学成分。因此,LYTAC 很有希望成为靶向细胞外蛋白和膜蛋白的通用平台,因为所有蛋白编码基因中有 40% 是细胞外蛋白和膜蛋白的产物。然而,对于癌症的靶向治疗,还需要考虑以下问题:需要全面的体内证据来支持 LYTAC 作为一种治疗选择的药效学、药代动力学和安全性;需要合作诊断来确定靶蛋白、预测性生物标志物和 LYTAC 相关调节剂的特征,以确认合适的癌症患者,并确定是否需要联合治疗,例如与 ALGO12 或 GNPTAB 抑制剂联合治疗;许多致癌膜蛋白的突变会导致肿瘤的异质性和/或耐药性,LYTAC 能否克服或绕过蛋白突变的负面影响尚不确定。自 2020 年以来,Lycia Therapeutics(由 Bertozzi 教授创立)和 Avilar Therapeutics 等公司围绕 LYTAC 技术开展了转化研究。我们期待这些新发现能推动LYTAC成为造福患者的新型疗法。郑清泉和蒲文臣提出了构思和大纲。郑清泉、郭嘉伟和蒲文臣组织并处理了图表。郭嘉伟和马锐修改了稿件。郭嘉伟和蒲文臣参与研究督导。所有作者均已阅读并批准最终稿件。作者声明无利益冲突。作者声明本亮点研究无需人类伦理批准。
{"title":"Lysosome-targeting chimera (LYTAC): A silver bullet for targeted degradation of oncogenic membrane proteins","authors":"Qingquan Zheng,&nbsp;Jiawei Guo,&nbsp;Rui Ma,&nbsp;Wenchen Pu","doi":"10.1002/mog2.64","DOIUrl":"https://doi.org/10.1002/mog2.64","url":null,"abstract":"<p>Recently, the group of Prof. Carolyn Bertozzi, a laureate of the Nobel Prize in chemistry 2022, reported the detailed mechanism of lysosome-targeting chimera (LYTAC) in the journal of <i>Science</i>,<span><sup>1</sup></span> after the publication of their first LYTAC molecule in <i>Nature</i> in 2020.<span><sup>2</sup></span> The establishment of LYTAC, a subtype of targeted protein degradation technology, expands the scope of protein degradation to extracellular and membrane-associated targets, and Bertozzi group's new discovery is expected to accelerate the development of LYTAC in cancer therapy.</p><p>Cell membranes play a critical role in various cellular processes, including signaling transduction, cell adhesion, transport of biomolecules and immunity. Proteins embedded in or associated with the cell membrane are key executants of the function of cell membrane, and their dysregulation contributes to tumorigenesis and development of human cancers.<span><sup>3</sup></span> For example, epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase for epithelial growth factor (EGF) and transforming growth factor α (TGF-α), belonging to the ErbB receptor family. Activation of EGFR signaling promotes cell proliferation, survival, angiogenesis and metastasis of diverse malignancies.<span><sup>3</sup></span> Moreover, hepatocyte growth factor receptor (c-Met, HGFR) is another oncogenic receptor tyrosine kinase in diverse cancers. Upon the binding to hepatocyte growth factor (HGF), c-Met is activated via autophosphorylation, leading to the initiation of oncogenic downstream signaling cascades, such as PI3K/AKT and RAS/ERK pathways.<span><sup>3</sup></span> Given their central role in cancer-promoting processes, EGFR and c-Met has become attractive targets for cancer therapies. Small-molecule tyrosine kinase inhibitors (EGFR: gefitinib, afatinib, osimertinib, etc.; c-Met: capmatinib, tepotinib, savolitinib, etc.) and monoclonal antibodies (EGFR: cetuximab, panitumumab; EGFR/c-Met: amivantamab; c-Met: emibetuzumab), have been developed and approved for the treatment of various cancers, including lung and colorectal cancers (Figure 1A). But severe acquired resistance (e.g., via EGFR mutations) and limited therapeutic efficacy (slightly prolonged overall survival) of these treatments restrict their clinical benefit for patients. Moreover, nonenzymatic function of membrane proteins, such as protein–protein interactions, could not be interfered with by kinase inhibitors or monoclonal antibodies, calling for new strategies to control these oncogenic membrane proteins.</p><p>Targeted protein degradation (TPD) is a therapeutic approach that aims to selectively remove disease-causing or undesirable proteins from cells by inducing their degradation, with multiple therapies entering clinical trials and targeting proteins that are previously considered “undruggable.”<span><sup>4</sup></span> There are two main protein degradation mechanisms within cel","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.64","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139468301","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 regulation in cancer therapy: From mechanisms to clinical advances 癌症治疗中的表观遗传调控:从机制到临床进展
Pub Date : 2024-01-04 DOI: 10.1002/mog2.59
Lei Tao, Yue Zhou, Yuan Luo, Jiahao Qiu, Yuzhou Xiao, Jiao Zou, Yu Zhang, Xingchen Liu, Xinyu Yang, Kun Gou, Jing Xu, Xinqi Guan, Xiaobo Cen, Yinglan Zhao

Epigenetic regulation refers to the alteration of gene expression independent of changes in DNA sequence. It involves chemical modifications such as DNA methylation, histone methylation, and histone acetylation, which are regulated by a coordinated interplay of various regulators to ensure precise spatial and temporal regulation of gene expression. Epigenetic aberrations are commonly observed in cancer and are considered as hallmarks of cancer. In recent years, small molecules targeting specific epigenetic regulators have been developed and are demonstrating promising therapeutic potential in preclinical and clinical trials for cancer treatment. In this review, we summarize the essential regulatory mechanisms and dysfunctions of epigenetic regulators involved in DNA methylation, histone methylation, and histone acetylation during tumor development and progression. Moreover, we discuss the current advances and challenges in cancer epigenetic therapy that target these mechanisms in both hematologic malignancies and solid tumors. Finally, we discuss the potential of combining epigenetic drugs with other therapies, including chemotherapy, radiotherapy, targeted therapy, and immunotherapy, as a promising approach for cancer treatment. Overall, we aim to enhance the understanding of epigenetic regulation in cancer therapy and explore targeted therapeutic strategies based on these mechanisms, to ultimately advance cancer therapy and improve patient prognosis.

表观遗传调控是指基因表达的改变与 DNA 序列的变化无关。它涉及 DNA 甲基化、组蛋白甲基化和组蛋白乙酰化等化学修饰,通过各种调节因子的协调相互作用来确保基因表达在空间和时间上的精确调控。表观遗传畸变常见于癌症,被认为是癌症的标志。近年来,针对特定表观遗传调控因子的小分子药物不断被开发出来,并在癌症治疗的临床前和临床试验中显示出良好的治疗潜力。在这篇综述中,我们总结了肿瘤发生和发展过程中 DNA 甲基化、组蛋白甲基化和组蛋白乙酰化所涉及的表观遗传调控因子的基本调控机制和功能障碍。此外,我们还讨论了目前针对血液系统恶性肿瘤和实体瘤这些机制的癌症表观遗传疗法的进展和挑战。最后,我们讨论了将表观遗传学药物与化疗、放疗、靶向治疗和免疫治疗等其他疗法相结合,作为一种有前途的癌症治疗方法的潜力。总之,我们的目标是加深对表观遗传调控在癌症治疗中的作用的理解,并根据这些机制探索靶向治疗策略,最终推进癌症治疗,改善患者预后。
{"title":"Epigenetic regulation in cancer therapy: From mechanisms to clinical advances","authors":"Lei Tao,&nbsp;Yue Zhou,&nbsp;Yuan Luo,&nbsp;Jiahao Qiu,&nbsp;Yuzhou Xiao,&nbsp;Jiao Zou,&nbsp;Yu Zhang,&nbsp;Xingchen Liu,&nbsp;Xinyu Yang,&nbsp;Kun Gou,&nbsp;Jing Xu,&nbsp;Xinqi Guan,&nbsp;Xiaobo Cen,&nbsp;Yinglan Zhao","doi":"10.1002/mog2.59","DOIUrl":"https://doi.org/10.1002/mog2.59","url":null,"abstract":"<p>Epigenetic regulation refers to the alteration of gene expression independent of changes in DNA sequence. It involves chemical modifications such as DNA methylation, histone methylation, and histone acetylation, which are regulated by a coordinated interplay of various regulators to ensure precise spatial and temporal regulation of gene expression. Epigenetic aberrations are commonly observed in cancer and are considered as hallmarks of cancer. In recent years, small molecules targeting specific epigenetic regulators have been developed and are demonstrating promising therapeutic potential in preclinical and clinical trials for cancer treatment. In this review, we summarize the essential regulatory mechanisms and dysfunctions of epigenetic regulators involved in DNA methylation, histone methylation, and histone acetylation during tumor development and progression. Moreover, we discuss the current advances and challenges in cancer epigenetic therapy that target these mechanisms in both hematologic malignancies and solid tumors. Finally, we discuss the potential of combining epigenetic drugs with other therapies, including chemotherapy, radiotherapy, targeted therapy, and immunotherapy, as a promising approach for cancer treatment. Overall, we aim to enhance the understanding of epigenetic regulation in cancer therapy and explore targeted therapeutic strategies based on these mechanisms, to ultimately advance cancer therapy and improve patient prognosis.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"3 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2024-01-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.59","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139109890","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
Cancer stem cells: Signaling pathways and therapeutic targeting 癌症干细胞:信号通路和靶向治疗
Pub Date : 2023-12-28 DOI: 10.1002/mog2.62
Joyeeta Talukdar, Tryambak P. Srivastava, Om S. Sahoo, Abhibroto Karmakar, Avdhesh K. Rai, Anupam Sarma, Gayatri Gogoi, Mohammed S. Alqahtani, Mohamed Abbas, Ruby Dhar, Subhradip Karmakar

Cancer stem cells (CSCs) constitute a minority cell population characterized by unbounded proliferative potential in both solid and hematological cancers. Despite sharing key stem cell attributes, CSCs possess unique traits, including the initiation and propagation of tumors and resistance to conventional therapies. The purpose of this review is to delve into the origins and fundamental characteristics of CSCs, emphasizing their role in tumor growth and metastasis. The focus extends to unraveling cellular signaling pathways driving oncogenic processes and understanding aberrant cellular crosstalk crucial for targeted cancer therapies. Beginning with an exploration of CSC properties and behavior, we progress to dissecting the cellular signaling network that fuels oncogenic pathways. The discussion spans the inception of CSCs, their survival strategies, and adaptation to new environments. We then transit to recent therapeutic advancements targeting CSCs, culminating in an exploration for precise therapeutic targeting. This review henceforth, underscores the vital significance of comprehending CSCs in cancer progression and treatment resistance. By unraveling the complex signaling pathways and survival mechanisms unique to CSCs, it paves the way for targeted therapeutic strategies that hold immense promise in enhancing cancer treatment efficacy while minimizing collateral damage.

癌症干细胞(CSCs)是一种少数细胞群,其特点是在实体癌和血癌中具有无限的增殖潜力。尽管共享关键的干细胞属性,但癌症干细胞具有独特的特征,包括肿瘤的诱发和繁殖以及对传统疗法的抗药性。本综述旨在深入探讨CSCs的起源和基本特征,强调它们在肿瘤生长和转移中的作用。重点还包括揭示驱动致癌过程的细胞信号通路,以及了解对癌症靶向疗法至关重要的异常细胞串联。我们从探讨 CSC 的特性和行为开始,进而剖析助长致癌途径的细胞信号网络。讨论涵盖了 CSC 的起源、生存策略和对新环境的适应。然后,我们转入针对 CSCs 的最新治疗进展,最后探讨了精确靶向治疗。这篇综述强调了了解 CSCs 在癌症进展和耐药性方面的重要意义。通过揭示 CSCs 独有的复杂信号通路和生存机制,它为靶向治疗策略铺平了道路,而靶向治疗策略在提高癌症治疗效果的同时又能最大限度地减少附带损害。
{"title":"Cancer stem cells: Signaling pathways and therapeutic targeting","authors":"Joyeeta Talukdar,&nbsp;Tryambak P. Srivastava,&nbsp;Om S. Sahoo,&nbsp;Abhibroto Karmakar,&nbsp;Avdhesh K. Rai,&nbsp;Anupam Sarma,&nbsp;Gayatri Gogoi,&nbsp;Mohammed S. Alqahtani,&nbsp;Mohamed Abbas,&nbsp;Ruby Dhar,&nbsp;Subhradip Karmakar","doi":"10.1002/mog2.62","DOIUrl":"https://doi.org/10.1002/mog2.62","url":null,"abstract":"<p>Cancer stem cells (CSCs) constitute a minority cell population characterized by unbounded proliferative potential in both solid and hematological cancers. Despite sharing key stem cell attributes, CSCs possess unique traits, including the initiation and propagation of tumors and resistance to conventional therapies. The purpose of this review is to delve into the origins and fundamental characteristics of CSCs, emphasizing their role in tumor growth and metastasis. The focus extends to unraveling cellular signaling pathways driving oncogenic processes and understanding aberrant cellular crosstalk crucial for targeted cancer therapies. Beginning with an exploration of CSC properties and behavior, we progress to dissecting the cellular signaling network that fuels oncogenic pathways. The discussion spans the inception of CSCs, their survival strategies, and adaptation to new environments. We then transit to recent therapeutic advancements targeting CSCs, culminating in an exploration for precise therapeutic targeting. This review henceforth, underscores the vital significance of comprehending CSCs in cancer progression and treatment resistance. By unraveling the complex signaling pathways and survival mechanisms unique to CSCs, it paves the way for targeted therapeutic strategies that hold immense promise in enhancing cancer treatment efficacy while minimizing collateral damage.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"2 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2023-12-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.62","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139101183","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
Application of machine learning to classify cancers of unknown primary 应用机器学习对原发灶不明的癌症进行分类
Pub Date : 2023-12-19 DOI: 10.1002/mog2.63
Shuvam Sarkar, Daniel T. Baptista-Hon

A recent study by Moon et al.1 published in Nature Medicine highlights the role of OncoNPC, a machine learning tool, in diagnosing cancers of unknown primary (CUP). The study offers an insight into the efficacy and accessibility of OncoNPC over traditional diagnostic tools and highlights the wider implications of machine learning technologies in delivering precision medicine.

The emergence of targeted immunotherapy over the past decade has led to a paradigm shift in clinical oncology. The efficacy of therapeutic agents in treating cancers such as chronic myeloid leukemia and HER2-positive breast cancer, for example, are now well established.2 However, CUPs, metastatic diseases where the primary tumor could not be identified present a significant challenge in this new era of precision medicine. CUPs account for 3%–5% of cancer diagnoses and present significant challenges in providing targeted therapy due to diagnostic uncertainty, and limited therapeutic targets.3 Indeed, the mortality rate in patients with CUPs is up to 80% at 12 months postdiagnosis.4

Several authors have hypothesized pathological mechanisms that might underlie CUPs. Lopez-Lazaro5 reconciled existing research on stem cells driving tumorigenesis by suggesting CUPs may occur as a result of stem cell migration followed by malignant transformation. This could, in theory, present as metastatic cancer in the absence of a clear primary tumor. Alternative studies have suggested CUPs occur from early dissemination of a primary tumor resulting in rapidly progressive metastatic disease.4 This would account for the significant mortality rate associated with CUPs as early dissemination could increase metastatic burden and limit therapeutic interventions.

Current approaches for investigating CUPs focus primarily on immunohistochemistry (IHC) techniques or molecular profiling of tumor samples. Interpretation of IHC results can be inherently subjective. Studies using IHC techniques to investigate CUPs were only able to suggest a primary tumor in 25% of patients.6 Molecular profiling compromises several techniques such as whole genome sequencing or gene expression analysis to determine the primary tumor based on the molecular characteristics of tumor cells. The efficacy of these methods remains unclear, however, as implementation into clinical practice is often limited by cost-effectiveness.

Moon et al utilized next-generation sequencing (NGS) data within this study to guide genomic profiling of CUPs.1 NGS elicits a cellular genetic profile by simultaneously analyzing millions of fragments of DNA. This method is relatively cost-effective and significant tumor NGS data already exists.7 This study therefore uses NGS data in concordance with electronic health

Moon等人最近在《自然医学》(Nature Medicine)上发表的一项研究1 强调了机器学习工具OncoNPC在诊断原发性不明癌症(CUP)中的作用。该研究深入探讨了OncoNPC相对于传统诊断工具的有效性和可及性,并强调了机器学习技术在提供精准医疗方面的广泛意义。例如,治疗慢性骨髓性白血病和 HER2 阳性乳腺癌等癌症的药物疗效现已得到公认。2 然而,在这个精准医疗的新时代,无法确定原发肿瘤的转移性疾病 CUPs 面临着巨大的挑战。CUPs 占癌症诊断的 3%-5%,由于诊断不确定性和治疗靶点有限,给靶向治疗带来了巨大挑战。洛佩兹-拉扎罗5(Lopez-Lazaro5)调和了干细胞驱动肿瘤发生的现有研究,认为银联可能是干细胞迁移后恶性转化的结果。理论上,这可能在没有明确原发肿瘤的情况下表现为转移性癌症。目前研究 CUP 的方法主要集中于免疫组化(IHC)技术或肿瘤样本的分子图谱分析。对 IHC 结果的解释可能具有固有的主观性。使用 IHC 技术调查 CUP 的研究仅能提示 25% 的患者为原发性肿瘤。6 分子图谱分析综合了多种技术,如全基因组测序或基因表达分析,以根据肿瘤细胞的分子特征确定原发性肿瘤。Moon 等人在这项研究中利用了新一代测序(NGS)数据来指导 CUPs 的基因组图谱分析1。1 NGS 通过同时分析数百万个 DNA 片段来绘制细胞基因图谱,这种方法成本效益相对较高,而且已经存在大量肿瘤 NGS 数据。7 因此,本研究利用 NGS 数据和电子健康记录来回顾性预测 971 例 CUP 患者的原发性肿瘤。作者开发了一种新型机器学习工具 OncoNPC,该工具是在已知原发肿瘤类型患者的 NGS 数据上训练出来的。OncoNPC 能够以较高的置信度对已知原发肿瘤患者的 22 种癌症类型进行分类,并考虑到了患者人口统计学特征的变化。有趣的是,与罕见组别相比,常见癌症亚型的识别准确率更高。然后将 OncoNPC 应用于 CUP 患者,结果以高置信度预测出 41% 患者的原发性癌症。这表明有很大一部分 CUP 是罕见肿瘤。最常见的原发肿瘤是肺癌、胰腺癌和肠癌,这与已有的 CUP 死亡病例尸检数据一致8。因此,与 IHC 等现有技术相比,OncoNPC 可以提供更客观的 CUP 分析方法。无论用户经验如何,都能预测原发亚型和相关置信区间。此外,一旦根据基线数据对该工具进行了培训,临床应用就不会耗费大量资源,因此比 IHC 或分子图谱分析更容易获得。根据种系变异数据计算了多基因风险评分,发现与已知原发性癌症患者相比,CUP 患者的种系风险更大。OncoNPC 还能根据预测的癌症亚型进行风险分层,其中胃癌和胰腺癌的预后最差。对 158 例接受姑息治疗的 CUP 患者进行的回顾性分析发现,与 CUP 肿瘤亚型一致的治疗可显著改善患者的生存预后。值得注意的是,OncoNPC 在这一队列中又发现了 24 名患者,他们可能适合在姑息治疗前接受基因组靶向治疗。这项研究让人们深入了解了机器学习工具在促进个性化医疗的出现以及确定 CUP 患者潜在治疗靶点方面的作用。
{"title":"Application of machine learning to classify cancers of unknown primary","authors":"Shuvam Sarkar,&nbsp;Daniel T. Baptista-Hon","doi":"10.1002/mog2.63","DOIUrl":"https://doi.org/10.1002/mog2.63","url":null,"abstract":"<p>A recent study by Moon et al.<span><sup>1</sup></span> published in Nature Medicine highlights the role of OncoNPC, a machine learning tool, in diagnosing cancers of unknown primary (CUP). The study offers an insight into the efficacy and accessibility of OncoNPC over traditional diagnostic tools and highlights the wider implications of machine learning technologies in delivering precision medicine.</p><p>The emergence of targeted immunotherapy over the past decade has led to a paradigm shift in clinical oncology. The efficacy of therapeutic agents in treating cancers such as chronic myeloid leukemia and HER2-positive breast cancer, for example, are now well established.<span><sup>2</sup></span> However, CUPs, metastatic diseases where the primary tumor could not be identified present a significant challenge in this new era of precision medicine. CUPs account for 3%–5% of cancer diagnoses and present significant challenges in providing targeted therapy due to diagnostic uncertainty, and limited therapeutic targets.<span><sup>3</sup></span> Indeed, the mortality rate in patients with CUPs is up to 80% at 12 months postdiagnosis.<span><sup>4</sup></span></p><p>Several authors have hypothesized pathological mechanisms that might underlie CUPs. Lopez-Lazaro<span><sup>5</sup></span> reconciled existing research on stem cells driving tumorigenesis by suggesting CUPs may occur as a result of stem cell migration followed by malignant transformation. This could, in theory, present as metastatic cancer in the absence of a clear primary tumor. Alternative studies have suggested CUPs occur from early dissemination of a primary tumor resulting in rapidly progressive metastatic disease.<span><sup>4</sup></span> This would account for the significant mortality rate associated with CUPs as early dissemination could increase metastatic burden and limit therapeutic interventions.</p><p>Current approaches for investigating CUPs focus primarily on immunohistochemistry (IHC) techniques or molecular profiling of tumor samples. Interpretation of IHC results can be inherently subjective. Studies using IHC techniques to investigate CUPs were only able to suggest a primary tumor in 25% of patients.<span><sup>6</sup></span> Molecular profiling compromises several techniques such as whole genome sequencing or gene expression analysis to determine the primary tumor based on the molecular characteristics of tumor cells. The efficacy of these methods remains unclear, however, as implementation into clinical practice is often limited by cost-effectiveness.</p><p>Moon et al utilized next-generation sequencing (NGS) data within this study to guide genomic profiling of CUPs.<span><sup>1</sup></span> NGS elicits a cellular genetic profile by simultaneously analyzing millions of fragments of DNA. This method is relatively cost-effective and significant tumor NGS data already exists.<span><sup>7</sup></span> This study therefore uses NGS data in concordance with electronic health ","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"2 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2023-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.63","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138739871","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
Cellular senescence: A potential mode of circular RNAs regulating prostate cancer 细胞衰老:环状核糖核酸调控前列腺癌的潜在模式
Pub Date : 2023-12-14 DOI: 10.1002/mog2.61
Yunpeng Li, Aoyu Fan, Yunyan Zhang, Ziyi Guo, Wei Meng, Wei Pan, Zhongliang Ma, Wei Chen

Cellular senescence is a state characterized by permanent cell cycle stoppage, which has long been viewed as a protective mechanism against neoplasia. However, accumulating evidence reveal that cellular senescence variously stimulates tumorigenesis and malignant progression in certain contexts. Senescence-associated secretory phenotype (SASP) is a crucial feature of senescent cells and the main way they function. Prostate cancer (PCa) is apparently an age-related tumor with a high prevalence in the elderly. With the aggravation of population aging the morbidity of PCa continues to rise. And with the progress of the disease, most patients eventually develop castration-resistant PCa (CRPC) or drug resistance, which poses a challenge for the treatment of PCa and aggravates the burden on patients and society. Circular RNAs (circRNAs) are a class of endogenous noncoding RNAs formed by back-splicing of pre-mRNAs. Characterized by special covalently closed circular structure, they play important regulatory roles in various tumors. Numerous studies have revealed that circRNAs can regulate PCa and cellular senescence in diverse ways. This review explores a potential mode that circRNAs regulate PCa, reveales a significant mechanism of tumorigenesis and progression for PCa, suggesting a new strategy for PCa research.

细胞衰老是一种以细胞周期永久停止为特征的状态,长期以来被认为是对抗肿瘤的一种保护机制。然而,越来越多的证据表明,在某些情况下,细胞衰老以不同的方式刺激肿瘤发生和恶性进展。衰老相关分泌表型(senescence associated secretory phenotype, SASP)是衰老细胞的一个重要特征,也是衰老细胞发挥功能的主要途径。前列腺癌(PCa)显然是一种与年龄相关的肿瘤,在老年人中发病率很高。随着人口老龄化的加剧,前列腺癌的发病率持续上升。并且随着病情的发展,大多数患者最终发展为去势抵抗性PCa (CRPC)或耐药,这给PCa的治疗带来了挑战,加重了患者和社会的负担。环状rna (circRNAs)是一类内源性非编码rna,由前mrna的反剪接形成。它们具有共价封闭的特殊环状结构,在多种肿瘤中发挥重要的调节作用。大量研究表明,环状rna可以以多种方式调节PCa和细胞衰老。本文探讨了环状rna调控前列腺癌的潜在模式,揭示了前列腺癌发生发展的重要机制,为前列腺癌研究提供了新的策略。
{"title":"Cellular senescence: A potential mode of circular RNAs regulating prostate cancer","authors":"Yunpeng Li,&nbsp;Aoyu Fan,&nbsp;Yunyan Zhang,&nbsp;Ziyi Guo,&nbsp;Wei Meng,&nbsp;Wei Pan,&nbsp;Zhongliang Ma,&nbsp;Wei Chen","doi":"10.1002/mog2.61","DOIUrl":"https://doi.org/10.1002/mog2.61","url":null,"abstract":"<p>Cellular senescence is a state characterized by permanent cell cycle stoppage, which has long been viewed as a protective mechanism against neoplasia. However, accumulating evidence reveal that cellular senescence variously stimulates tumorigenesis and malignant progression in certain contexts. Senescence-associated secretory phenotype (SASP) is a crucial feature of senescent cells and the main way they function. Prostate cancer (PCa) is apparently an age-related tumor with a high prevalence in the elderly. With the aggravation of population aging the morbidity of PCa continues to rise. And with the progress of the disease, most patients eventually develop castration-resistant PCa (CRPC) or drug resistance, which poses a challenge for the treatment of PCa and aggravates the burden on patients and society. Circular RNAs (circRNAs) are a class of endogenous noncoding RNAs formed by back-splicing of pre-mRNAs. Characterized by special covalently closed circular structure, they play important regulatory roles in various tumors. Numerous studies have revealed that circRNAs can regulate PCa and cellular senescence in diverse ways. This review explores a potential mode that circRNAs regulate PCa, reveales a significant mechanism of tumorigenesis and progression for PCa, suggesting a new strategy for PCa research.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"2 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2023-12-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.61","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138634351","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
Novel insights into HBV-hepatocellular carcinoma at single-cell sequencing 单细胞测序对 HBV 肝细胞癌的新认识
Pub Date : 2023-12-11 DOI: 10.1002/mog2.60
Dandan Yin, Tao Zhou, Xuyang Xia, Chang Han, Zhaoqian Liu, Qiu Li, Yang Shu, Heng Xu

A significant proportion of hepatocellular carcinoma (HCC) is pathologically associated with hepatitis B virus (HBV) infection, followed by unsatisfied clinical outcomes. The increasing unmet need for HBV-associated hepatocellular carcinoma (HBV-HCC) treatment drives to deeper understand the role of the intricate immune microenvironment, tumor cell plasticity and dynamics of tumor evolution in HBV-associated hepatic carcinogenesis. Thus, a comprehensive understanding of cross-talk between HBV, host cells, and tumor microenvironment is of fundamental importance for identifying immune imbalance and heterogeneity in HBV-HCC. Over the past 5 years, the application of single-cell RNA sequencing (scRNA-seq) in the understanding of heterogeneity and dynamics of immune cells, clonal evolution, and cancer stem cell (CSC) subsets of tumor cells has established a landscape for HBV-HCC tumor ecosystem. Novel insights into anatomizing immune escape mechanisms and tumor drug resistance have remarkably facilitated the revolution of HBV-HCC clinical treatment. Here, we provided a summary of HCC at single-cell resolution and details on the basic workflow, limitations, and improvements of scRNA-seq. The review highlights novel insights derived from scRNA-seq on advances in the immune microenvironment and tumor heterogeneity of HBV-HCC.

相当一部分肝细胞癌(HCC)在病理上与乙型肝炎病毒(HBV)感染有关,其临床结果也不尽如人意。HBV 相关肝细胞癌(HBV-HCC)治疗需求的不断增长促使人们更深入地了解错综复杂的免疫微环境、肿瘤细胞可塑性和肿瘤演变动态在 HBV 相关肝癌发生中的作用。因此,全面了解 HBV、宿主细胞和肿瘤微环境之间的交叉对话对于识别 HBV-HCC 中的免疫失衡和异质性至关重要。过去 5 年中,单细胞 RNA 测序(scRNA-seq)在了解免疫细胞的异质性和动态、克隆进化和肿瘤细胞的癌症干细胞(CSC)亚群方面的应用,为 HBV-HCC 肿瘤生态系统建立了一个图景。对免疫逃逸机制和肿瘤耐药性的新认识极大地推动了 HBV-HCC 临床治疗的变革。在此,我们对单细胞分辨率的 HCC 进行了总结,并详细介绍了 scRNA-seq 的基本工作流程、局限性和改进之处。综述重点介绍了 scRNA-seq 对 HBV-HCC 免疫微环境和肿瘤异质性进展的新见解。
{"title":"Novel insights into HBV-hepatocellular carcinoma at single-cell sequencing","authors":"Dandan Yin,&nbsp;Tao Zhou,&nbsp;Xuyang Xia,&nbsp;Chang Han,&nbsp;Zhaoqian Liu,&nbsp;Qiu Li,&nbsp;Yang Shu,&nbsp;Heng Xu","doi":"10.1002/mog2.60","DOIUrl":"https://doi.org/10.1002/mog2.60","url":null,"abstract":"<p>A significant proportion of hepatocellular carcinoma (HCC) is pathologically associated with hepatitis B virus (HBV) infection, followed by unsatisfied clinical outcomes. The increasing unmet need for HBV-associated hepatocellular carcinoma (HBV-HCC) treatment drives to deeper understand the role of the intricate immune microenvironment, tumor cell plasticity and dynamics of tumor evolution in HBV-associated hepatic carcinogenesis. Thus, a comprehensive understanding of cross-talk between HBV, host cells, and tumor microenvironment is of fundamental importance for identifying immune imbalance and heterogeneity in HBV-HCC. Over the past 5 years, the application of single-cell RNA sequencing (scRNA-seq) in the understanding of heterogeneity and dynamics of immune cells, clonal evolution, and cancer stem cell (CSC) subsets of tumor cells has established a landscape for HBV-HCC tumor ecosystem. Novel insights into anatomizing immune escape mechanisms and tumor drug resistance have remarkably facilitated the revolution of HBV-HCC clinical treatment. Here, we provided a summary of HCC at single-cell resolution and details on the basic workflow, limitations, and improvements of scRNA-seq. The review highlights novel insights derived from scRNA-seq on advances in the immune microenvironment and tumor heterogeneity of HBV-HCC.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"2 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2023-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.60","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138570982","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
COVID-19 and cancer: Dichotomy of the menacing dilemma COVID-19和癌症:威胁困境的二分法
Pub Date : 2023-11-30 DOI: 10.1002/mog2.58
Mrinal K. Ghosh, Shaheda Tabassum, Malini Basu

The coronavirus disease 2019 (COVID-19) pandemic brought about unprecedented challenges to global healthcare systems. Among the most vulnerable populations are cancer patients, who face dilemmas due to their compromised immune systems and the intricate interplay with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus. This comprehensive review delves into the multifaceted relationship between COVID-19 and cancer. Through an analysis of existing literature and clinical data, this review unravels the structural intricacies of the virus and examines its profound implications for cancer patients, thereby bridging the knowledge gap between virology and oncology. The review commences with an introduction regarding the COVID-19 pandemic and cancer. It then transitions into a detailed examination of the SARS-CoV-2 virus and its variants such as Alpha (PANGO lineage B.1.1.7), Beta (B.1.351), Gamma (P.1), Delta (B.1.617.2), and Omicron (B.1.1.529 lineage). Subsequently, an insightful analysis of the impact of COVID-19 on major cancer types (viz., Lung, Colon, Brain, and gastrointestinal cancer) is elaborated. Finally, the therapeutic avenues, oncological care, and management are discussed. The nexus between COVID-19 and cancer adds a layer of complexity to patient care, emphasizing the importance of tailored approaches for those grappling with both conditions. Amid the landscape defined by the evolving viral strains, this review navigates through the multifaceted implications of COVID-19 on cancer patients and underscores the significance of integrating virology and oncology.

2019冠状病毒病(COVID-19)大流行给全球卫生保健系统带来了前所未有的挑战。最脆弱的人群是癌症患者,由于免疫系统受损以及与严重急性呼吸综合征冠状病毒2 (SARS-CoV-2)病毒的复杂相互作用,他们面临困境。这篇综合综述深入探讨了COVID-19与癌症之间的多方面关系。通过对现有文献和临床数据的分析,本综述揭示了病毒结构的复杂性,并研究了其对癌症患者的深远影响,从而弥合了病毒学和肿瘤学之间的知识差距。本次审查首先介绍了COVID-19大流行和癌症。然后,它过渡到对SARS-CoV-2病毒及其变体的详细检查,如Alpha (PANGO谱系B.1.1.7)、Beta (B.1.351)、Gamma (P.1)、Delta (B.1.617.2)和Omicron (B.1.1.529谱系)。随后,深入分析了COVID-19对主要癌症类型(肺癌、结肠癌、脑癌和胃肠道癌)的影响。最后,讨论了治疗途径、肿瘤护理和管理。COVID-19和癌症之间的联系为患者护理增加了一层复杂性,强调了为同时患有这两种疾病的患者量身定制方法的重要性。在不断演变的病毒株所定义的环境中,本综述通过COVID-19对癌症患者的多方面影响进行了导航,并强调了整合病毒学和肿瘤学的重要性。
{"title":"COVID-19 and cancer: Dichotomy of the menacing dilemma","authors":"Mrinal K. Ghosh,&nbsp;Shaheda Tabassum,&nbsp;Malini Basu","doi":"10.1002/mog2.58","DOIUrl":"https://doi.org/10.1002/mog2.58","url":null,"abstract":"<p>The coronavirus disease 2019 (COVID-19) pandemic brought about unprecedented challenges to global healthcare systems. Among the most vulnerable populations are cancer patients, who face dilemmas due to their compromised immune systems and the intricate interplay with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus. This comprehensive review delves into the multifaceted relationship between COVID-19 and cancer. Through an analysis of existing literature and clinical data, this review unravels the structural intricacies of the virus and examines its profound implications for cancer patients, thereby bridging the knowledge gap between virology and oncology. The review commences with an introduction regarding the COVID-19 pandemic and cancer. It then transitions into a detailed examination of the SARS-CoV-2 virus and its variants such as Alpha (PANGO lineage B.1.1.7), Beta (B.1.351), Gamma (P.1), Delta (B.1.617.2), and Omicron (B.1.1.529 lineage). Subsequently, an insightful analysis of the impact of COVID-19 on major cancer types (viz., Lung, Colon, Brain, and gastrointestinal cancer) is elaborated. Finally, the therapeutic avenues, oncological care, and management are discussed. The nexus between COVID-19 and cancer adds a layer of complexity to patient care, emphasizing the importance of tailored approaches for those grappling with both conditions. Amid the landscape defined by the evolving viral strains, this review navigates through the multifaceted implications of COVID-19 on cancer patients and underscores the significance of integrating virology and oncology.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"2 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2023-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.58","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138468424","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
Systematic analysis of markers for T-cell differentiation revealing CD95 as a novel biomarker for prognosis and immunotherapeutic efficacy in colon cancer t细胞分化标志物的系统分析揭示CD95作为结肠癌预后和免疫治疗疗效的新生物标志物
Pub Date : 2023-11-19 DOI: 10.1002/mog2.57
Yuxin Shi, Jie Mei, Rui Hou, Hao Wang, Junli Ding, Junying Xu

Colon cancer is the third most frequently diagnosed cancer worldwide. Considerable progress has been made in the therapeutic strategies and the accuracy of predicting the patients' prognosis. In the past decade, immunotherapy, represented by programmed cell death 1 (PD-1) and programmed cell death 1 ligand 1 (PD-L1) signaling inhibitors have made remarkable advances in many solid tumors, but limited effect in colon cancer. In particular, colon cancer demonstrates a remarkably poor response to immunotherapy compared with other cancers, with a notable exception of tumors harboring high microsatellite instability (MSI-H) or deficient mismatch repair (dMMR).1 MSI-H or dMMR tumors are characterized by high mutational/neo-antigen burden, and an inflammatory tumor microenvironment with abundant tumor-infiltrating lymphocytes (TILs).2 Although better efficacy could be achieved in a small group of patients with MSI-H or dMMR tumors, a proportion of other patients could benefit from immunotherapy. Thus, more appropriate biomarkers are needed to be discovered to predict the patients' prognosis and immunotherapeutic efficacy.

Different T-cell subsets in the tumor immune microenvironment (TIME) are closely related to the antitumor response and the prognosis of tumor patients. Tumor-specific CD8+ T cells are the core cellular components that exert antitumor effects in TIME, by recognizing specific receptors on the tumor surface and secreting cytokines or via the Fas/FsaL pathway. Multiple T-cell subsets are characterized by respective differentiation markers. Currently, with the deepening research of T-cell subsets, the detection of relevant immune indicators and the formulation of therapeutic schedules have attracted increasing clinical attention. By monitoring the expression of T-cell differentiation markers in patients with colon cancer, the benefit of patients receiving immunotherapy can be further evaluated, thus providing a theoretical basis for judging the clinical prognosis and designing more feasible treatment plans.

In this research, we systematically analyzed the prognostic values of multiple markers for different T-cell subsets3 using the Cancer Genome Atlas (TCGA) data set. Among all markers for different T cell subsets, we found that CD95 expression was correlated with better overall survival (OS) and progression-free survival (PFS), while BCL2 and CD122 were only correlated with PFS (Figure 1A,B). Therefore, CD95 was speculated to be associated with prognosis in colon cancer and was selected for further investigation and validation. The analysis of single-cell RNA-sequencing data set confirmed that CD95 was highly expressed in immune cells, especially in various T-cell subtypes (Figure S1A–C). CD95 is a member of the tumor necrosis factor receptor (TNF-R) superfamily and treated as characteristic marker of memeroy cells. It could also be used as the

结肠癌是世界上第三大最常见的癌症。在治疗策略和预测患者预后的准确性方面取得了长足的进步。在过去的十年中,以程序性细胞死亡1 (PD-1)和程序性细胞死亡1配体1 (PD-L1)信号抑制剂为代表的免疫治疗在许多实体肿瘤中取得了显著进展,但在结肠癌中的效果有限。特别是,与其他癌症相比,结肠癌对免疫治疗的反应明显较差,但具有高微卫星不稳定性(MSI-H)或错配修复缺陷(dMMR)的肿瘤除外MSI-H或dMMR肿瘤的特点是高突变/新抗原负荷,肿瘤微环境具有丰富的肿瘤浸润淋巴细胞(til)虽然在一小部分MSI-H或dMMR肿瘤患者中可以获得更好的疗效,但也有一部分其他患者可以从免疫治疗中获益。因此,需要发现更合适的生物标志物来预测患者的预后和免疫治疗效果。肿瘤免疫微环境(TIME)中不同的t细胞亚群与肿瘤患者的抗肿瘤反应和预后密切相关。肿瘤特异性CD8+ T细胞通过识别肿瘤表面的特异性受体并分泌细胞因子或通过Fas/FsaL通路,是TIME中发挥抗肿瘤作用的核心细胞成分。多个t细胞亚群由各自的分化标记物表征。目前,随着t细胞亚群研究的不断深入,相关免疫指标的检测和治疗方案的制定越来越受到临床的重视。通过监测结肠癌患者t细胞分化标志物的表达,可以进一步评估患者接受免疫治疗的获益,从而为判断临床预后和设计更可行的治疗方案提供理论依据。在这项研究中,我们使用癌症基因组图谱(TCGA)数据集系统地分析了多个标记物对不同t细胞亚群的预后价值。在不同T细胞亚群的所有标记物中,我们发现CD95表达与更好的总生存期(OS)和无进展生存期(PFS)相关,而BCL2和CD122仅与PFS相关(图1A,B)。因此,我们推测CD95与结肠癌的预后相关,并选择CD95进行进一步的研究和验证。单细胞rna测序数据集分析证实,CD95在免疫细胞中高度表达,特别是在各种t细胞亚型中(图S1A-C)。CD95是肿瘤坏死因子受体(TNF-R)超家族的成员,被认为是记忆细胞的特征标志物。它也可以作为诱导细胞凋亡的最经典的死亡受体。CD95诱导的细胞凋亡途径是免疫系统清除病毒感染细胞或肿瘤细胞的有力途径,因此CD95最初被确定为肿瘤抑制基因。接下来,为了了解CD95在结肠癌中的潜在生物学作用,我们在TCGA数据集中应用了基因集富集分析(GSEA)。结果显示,CD95与多种重要的免疫相关过程显著相关,如同种异体移植排斥反应、炎症反应和干扰素- γ反应(图1C和表S1)。鉴于CD95与结肠癌免疫相关过程之间的密切相关性,我们接下来评估了CD95与综合免疫调节剂(包括趋化因子、受体、MHC分子、免疫抑制剂和免疫刺激剂)之间的相关性,这些免疫调节剂与TIME特征和免疫治疗反应相关。结果显示,CD95与大多数免疫调节剂呈正相关(图1D和表S2)。此外,我们还分析了CD95与ImmuCellAI工具计算的T细胞亚群之间的相关性,结果显示CD95与细胞毒性T细胞之间存在最强的正相关性(表S3)。所有的研究结果都表明CD95与发炎的TIME高度相关。特别是,大量证据表明,与熟练MMR (pMMR)患者相比,MSI-H/dMMR结肠癌患者从免疫治疗中获益更多,且免疫治疗效果乐观。随后,我们使用了三个经过验证的队列,即TCGA队列、GSE39582队列和CPTAC2队列,结果表明CD95在MSI-H/dMMR状态的肿瘤中上调(图S2A-C)。此外,还评估了CD95与几种与免疫治疗反应相关的常见突变(包括NF1、POLE、ARID1A和BRAF)之间的相关性。结果显示,基因突变的肿瘤表达更高的CD95(图S3A-D)。 此外,招募了一个由29个接受免疫治疗的结肠癌标本组成的小规模免疫治疗队列,以进一步验证CD95表达与免疫治疗反应之间的关联。免疫荧光显示CD95在免疫细胞和肿瘤细胞中均有表达(图1E)。考虑到CD95在t细胞分化中的作用,我们只计算CD95阳性的免疫细胞。结果表明,免疫治疗反应良好的患者与反应较差的患者相比,CD95表达较高(图1E)。最后,生存分析显示,CD95+免疫细胞浸润高的患者比CD95+免疫细胞浸润低的患者预后好(图1F)。综上所述,这些结果支持CD95可以作为一种新的有前景的生物标志物,对结肠癌的免疫治疗有益。CD95和CD95L在结肠癌中广泛表达,以往在结肠癌中的研究表明,将CD95信号从凋亡转换为非凋亡的ERK/NF-κB信号有利于肿瘤的发生、局部生长和向肝脏转移的能力,这些都依赖于CD95L的表达。此外,已有证据表明CD95主要实现NF-κB、MAPK和PI3K等非凋亡信号通路,参与细胞迁移、分化、存活和细胞因子分泌。不仅在结肠癌中,在肺癌、胶质母细胞瘤、卵巢癌和肝癌中,CD95在某些情况下的表达反过来激活其非凋亡通路,在肿瘤的发生和进展中发挥重要作用此外,最新的研究表明,靶向CD95在黑色素瘤中增加了细胞毒性T细胞和树突状细胞的水平我们假设CD95可能介导肿瘤细胞的免疫原性死亡。然而,CD95对肿瘤生物学行为的具体影响及其与免疫治疗疗效的相关性尚不清楚。基于上述发现CD95与TIME中多个免疫相关过程和免疫调节剂呈正相关,我们推测在结肠癌中CD95与炎症性TIME相关,并鉴定出有利于抗肿瘤免疫的免疫热肿瘤。此外,我们还发现CD95在MSI-H/dMMR状态的肿瘤中表达上调。具有dMMR状态的结肠肿瘤与较高的CD8+ TILs密度和较高的肿瘤突变负荷(TMB)相关,这些特性似乎是免疫治疗反应的生物标志物,并显示出积极的预后价值。考虑到结肠癌的高发病率和死亡率以及难以获得令人满意的治疗结果,需要CD95作为一种合适的生物标志物来帮助识别可能受益于免疫治疗的候选人,并可以评估临床结果和治疗反应,这可能弥补现有免疫治疗策略的不足。总之,目前的研究确定了潜在的生物标志物来预测结肠癌的临床结果和对免疫治疗的反应。经系统分析,CD95作为T细胞的记忆标记物,在临床应用中可能是预测预后和免疫治疗反应的合适、准确的生物标志物。科学地说,新的预测性生物标志物的鉴定将极大地扩展我们对结肠癌免疫机制的理解,为个体治疗策略的决策提供信息。史宇昕:数据策展(equal);形式分析(相等);方法(平等);资源(平等);可视化(平等);写作-原稿(同等)。杰梅:数据管理(平等);形式分析(相等);调查(平等);方法(平等);验证(平等);写作-原稿(同等)。侯睿:形式分析(支持);验证(支持)。王浩:形式分析(支持);资源(支持)。丁俊丽:概念化(平等);资金获取(支持);项目管理(同等);写作—评审与编辑(同等)。许俊英:概念化(平等);获得资金(牵头);项目管理(同等);写作—评审与编辑(同等)。所有作者都阅读并批准了最终稿件。作者声明无利益冲突。经南京医科大学临床研究伦理委员会(2022-383)伦理批准。这是一项回顾性研究,不需要试验注册号。所有参与者均获得书面知情同意。
{"title":"Systematic analysis of markers for T-cell differentiation revealing CD95 as a novel biomarker for prognosis and immunotherapeutic efficacy in colon cancer","authors":"Yuxin Shi,&nbsp;Jie Mei,&nbsp;Rui Hou,&nbsp;Hao Wang,&nbsp;Junli Ding,&nbsp;Junying Xu","doi":"10.1002/mog2.57","DOIUrl":"https://doi.org/10.1002/mog2.57","url":null,"abstract":"<p>Colon cancer is the third most frequently diagnosed cancer worldwide. Considerable progress has been made in the therapeutic strategies and the accuracy of predicting the patients' prognosis. In the past decade, immunotherapy, represented by programmed cell death 1 (PD-1) and programmed cell death 1 ligand 1 (PD-L1) signaling inhibitors have made remarkable advances in many solid tumors, but limited effect in colon cancer. In particular, colon cancer demonstrates a remarkably poor response to immunotherapy compared with other cancers, with a notable exception of tumors harboring high microsatellite instability (MSI-H) or deficient mismatch repair (dMMR).<span><sup>1</sup></span> MSI-H or dMMR tumors are characterized by high mutational/neo-antigen burden, and an inflammatory tumor microenvironment with abundant tumor-infiltrating lymphocytes (TILs).<span><sup>2</sup></span> Although better efficacy could be achieved in a small group of patients with MSI-H or dMMR tumors, a proportion of other patients could benefit from immunotherapy. Thus, more appropriate biomarkers are needed to be discovered to predict the patients' prognosis and immunotherapeutic efficacy.</p><p>Different T-cell subsets in the tumor immune microenvironment (TIME) are closely related to the antitumor response and the prognosis of tumor patients. Tumor-specific CD8<sup>+</sup> T cells are the core cellular components that exert antitumor effects in TIME, by recognizing specific receptors on the tumor surface and secreting cytokines or via the Fas/FsaL pathway. Multiple T-cell subsets are characterized by respective differentiation markers. Currently, with the deepening research of T-cell subsets, the detection of relevant immune indicators and the formulation of therapeutic schedules have attracted increasing clinical attention. By monitoring the expression of T-cell differentiation markers in patients with colon cancer, the benefit of patients receiving immunotherapy can be further evaluated, thus providing a theoretical basis for judging the clinical prognosis and designing more feasible treatment plans.</p><p>In this research, we systematically analyzed the prognostic values of multiple markers for different T-cell subsets<span><sup>3</sup></span> using the Cancer Genome Atlas (TCGA) data set. Among all markers for different T cell subsets, we found that CD95 expression was correlated with better overall survival (OS) and progression-free survival (PFS), while BCL2 and CD122 were only correlated with PFS (Figure 1A,B). Therefore, CD95 was speculated to be associated with prognosis in colon cancer and was selected for further investigation and validation. The analysis of single-cell RNA-sequencing data set confirmed that CD95 was highly expressed in immune cells, especially in various T-cell subtypes (Figure S1A–C). CD95 is a member of the tumor necrosis factor receptor (TNF-R) superfamily and treated as characteristic marker of memeroy cells. It could also be used as the","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"2 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2023-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.57","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138146306","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
Development of covalent inhibitors: Principle, design, and application in cancer 共价抑制剂的发展:原理、设计和在癌症中的应用
Pub Date : 2023-10-31 DOI: 10.1002/mog2.56
Lang Zheng, Yang Li, Defa Wu, Huan Xiao, Shilong Zheng, Guan Wang, Qiu Sun

Covalent inhibitors have been a rapidly growing field in drug discovery due to their therapeutic potential and unique advantages in cancer therapy. As opposed to noncovalent inhibitory drugs, covalent inhibitors reversibly or irreversibly modify proximal nucleophilic amino acid residues on proteins, aiming to selectively recognize and bind to protein targets and addressing some of the challenges faced by noncovalent drugs. Most successful targeted covalent inhibitors depend primarily on binding-site cysteine residues, but this has limitations for certain protein targets that lack targetable cysteine residues. Recently, the rational design of covalent inhibitors or covalent probes targeting other nucleophilic residues, such as lysine, tyrosine, serine, has turned out to be another promising strategy for cancer therapy. Thus, the development of novel strategies to extend the scope of covalent binding and improve the binding properties is required. This review gives a summary of the development of covalent inhibitors targeting noncysteine from different aspects, including target identification, structure–activity relationships, drug discovery strategies, and binding properties, in the hope of providing a scientific reference for future covalent drug discovery as a means of expanding research in cancer therapy.

共价抑制剂由于其治疗潜力和在癌症治疗中的独特优势,已成为药物发现的一个快速发展的领域。与非共价抑制药物相反,共价抑制剂可逆或不可逆地修饰蛋白质上的近端亲核氨基酸残基,旨在选择性地识别和结合蛋白质靶点,解决非共价药物面临的一些挑战。大多数成功的靶向共价抑制剂主要依赖于结合位点的半胱氨酸残基,但这对某些缺乏可靶向半胱氨酸残基的蛋白质靶标具有局限性。近年来,合理设计针对其他亲核残基(如赖氨酸、酪氨酸、丝氨酸)的共价抑制剂或共价探针已被证明是另一种有前景的癌症治疗策略。因此,需要开发新的策略来扩展共价结合的范围并改善其结合特性。本文从靶点鉴定、构效关系、药物发现策略、结合特性等方面综述了针对非半胱氨酸的共价抑制剂的研究进展,以期为今后共价药物的发现提供科学参考,从而扩大对肿瘤治疗的研究。
{"title":"Development of covalent inhibitors: Principle, design, and application in cancer","authors":"Lang Zheng,&nbsp;Yang Li,&nbsp;Defa Wu,&nbsp;Huan Xiao,&nbsp;Shilong Zheng,&nbsp;Guan Wang,&nbsp;Qiu Sun","doi":"10.1002/mog2.56","DOIUrl":"10.1002/mog2.56","url":null,"abstract":"<p>Covalent inhibitors have been a rapidly growing field in drug discovery due to their therapeutic potential and unique advantages in cancer therapy. As opposed to noncovalent inhibitory drugs, covalent inhibitors reversibly or irreversibly modify proximal nucleophilic amino acid residues on proteins, aiming to selectively recognize and bind to protein targets and addressing some of the challenges faced by noncovalent drugs. Most successful targeted covalent inhibitors depend primarily on binding-site cysteine residues, but this has limitations for certain protein targets that lack targetable cysteine residues. Recently, the rational design of covalent inhibitors or covalent probes targeting other nucleophilic residues, such as lysine, tyrosine, serine, has turned out to be another promising strategy for cancer therapy. Thus, the development of novel strategies to extend the scope of covalent binding and improve the binding properties is required. This review gives a summary of the development of covalent inhibitors targeting noncysteine from different aspects, including target identification, structure–activity relationships, drug discovery strategies, and binding properties, in the hope of providing a scientific reference for future covalent drug discovery as a means of expanding research in cancer therapy.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"2 4","pages":""},"PeriodicalIF":0.0,"publicationDate":"2023-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.56","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"135928320","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