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
{"title":"Targeting iron–sulfur cluster assembly scaffold protein as a vulnerability in KRAS-activated pancreatic ductal adenocarcinoma","authors":"Huarui Cai, Hongjuan Cui, 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}
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
{"title":"Lysosome-targeting chimera (LYTAC): A silver bullet for targeted degradation of oncogenic membrane proteins","authors":"Qingquan Zheng, Jiawei Guo, Rui Ma, 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}
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, 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","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}
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.
{"title":"Cancer stem cells: Signaling pathways and therapeutic targeting","authors":"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","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}
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, 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}
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.
{"title":"Cellular senescence: A potential mode of circular RNAs regulating prostate cancer","authors":"Yunpeng Li, Aoyu Fan, Yunyan Zhang, Ziyi Guo, Wei Meng, Wei Pan, Zhongliang Ma, 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}
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.
{"title":"Novel insights into HBV-hepatocellular carcinoma at single-cell sequencing","authors":"Dandan Yin, Tao Zhou, Xuyang Xia, Chang Han, Zhaoqian Liu, Qiu Li, Yang Shu, 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}
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.
{"title":"COVID-19 and cancer: Dichotomy of the menacing dilemma","authors":"Mrinal K. Ghosh, Shaheda Tabassum, 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}
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
{"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, Jie Mei, Rui Hou, Hao Wang, Junli Ding, 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}
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.
{"title":"Development of covalent inhibitors: Principle, design, and application in cancer","authors":"Lang Zheng, Yang Li, Defa Wu, Huan Xiao, Shilong Zheng, Guan Wang, 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}