{"title":"Phosphatase Dysregulation in Cancer: Signaling Pathways and Therapeutic Opportunities","authors":"Maryam Jama, Michael Overduin, Khaled H. Barakat","doi":"10.1002/mog2.70028","DOIUrl":"https://doi.org/10.1002/mog2.70028","url":null,"abstract":"<p>Phosphatases are increasingly recognized as critical regulators of cancer biology, with important roles in both tumor cells and the tumor immune microenvironment (TIME). These enzymes modulate intracellular signaling pathways that control tumor growth, immune evasion, and metastasis. Although phosphatases were once considered undruggable, recent advances have highlighted their therapeutic potential. Despite growing evidence, phosphatases remain underexplored as drug targets, with no approved therapies to date. This review presents an in-depth overview of phosphatase classification based on catalytic domain similarities and explores their diverse functions as tumor suppressors, oncogenic drivers, or context-dependent regulators. We describe how phosphatases such as PTPN6, PTPN22, and DUSPs regulate key pathways like RAS/MAPK and PI3K/AKT in both tumor and immune cells. Additionally, we discuss the role of phosphatases in shaping the tumor microenvironment through exosome secretion. This review highlights current therapeutic strategies, including small molecules and antibodies, and their synergistic effects with kinase inhibitors and immune checkpoint blockade. By summarizing recent advances, this paper underscores the need for deeper mechanistic insights into phosphatase function in cancer and immunity. Understanding these mechanisms will be key to unlocking their potential as novel therapeutic targets in oncology.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 2","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-06-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70028","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144472805","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}
Xin Wang, Yiyao Zeng, Zihan Xu, Xiangyue Meng, Jie Chen
{"title":"A Causal Relationship Between Genetically Proxied Inhibition of HMGCR, NPC1L1, and PCSK9 and Cancers","authors":"Xin Wang, Yiyao Zeng, Zihan Xu, Xiangyue Meng, Jie Chen","doi":"10.1002/mog2.70026","DOIUrl":"https://doi.org/10.1002/mog2.70026","url":null,"abstract":"<p>Cancer remains a significant global health burden, ranking as the second leading cause of death despite considerable medical advances. Increasing evidence links cholesterol metabolism, particularly low-density lipoprotein cholesterol (LDL-C), to cancer risk. Epidemiological studies suggest that elevated LDL-C levels are associated with multiple cancers, including breast, colorectal, and pancreatic cancers [<span>1</span>]. These observations suggest that LDL-C-lowering therapies may have anticancer effects. Although preclinical studies show that statins can inhibit tumor growth and metastasis, the effects of different lipid-lowering agents on cancer risk remain unclear. To investigate this, we conducted a Mendelian randomization (MR) study leveraging genetically proxied LDL-C-lowering drug targets, including HMG-CoA reductase (HMGCR), proprotein convertase subtilisin/kexin type 9 (PCSK9), and Niemann-Pick C1-like 1 (NPC1L1) inhibitors. We examined the associations between these drug targets and 16 different cancer types, as well as overall cancer risk (Figure 1A).</p><p>Figure 1B presented the outcomes of MR analyses investigating the causal effects of genetically proxied lipid-lowering drug targets on 17 different types of cancer, alongside pleiotropy assessments. Funnel plots were used to evaluate heterogeneity. A rigorous instrument selection process yielded between 3 and 60 single nucleotide polymorphisms (SNPs) per target, with all F-statistics > 10, mitigating the risk of weak instrument bias. As for the PCSK9 inhibition (PCSK9i), the fixed-effect inverse-variance weighted (IVW) method clearly demonstrated a protective effect on breast cancer (OR: 0.9124, <i>p</i> = 0.0071, 95%CI: 0.8455–0.9792), which this result was corroborated by the weighted median and the weighted mode method. In addition, it also identified PCSK9i had an obvious protective effect on thyroid cancer (OR: 0.3272, <i>p</i> = 0.0397, 95%CI: 0.0370–0.5915) and brain cancer (OR: 0.9985, <i>p</i> = 1.8786E-07, 95%CI: 0.9980–0.9991), only in the IVW method. More importantly, the PCSK9i retained a strong protective association with overall cancer (OR: 0.9883, <i>p</i> = 5.8596E-11, 95%CI: 0.9848–0.9918). However, PCSK9i showed significant associations with higher risk of oesophageal cancer (OR: 1.0009, <i>p</i> = 0.0028, 95%CI: 1.0003–1.0014) and lung cancer (OR: 1.0328, <i>p</i> = 0.0020, 95%CI: 1.0054–1.2782). Additionally, genetically predicted HMGCR inhibition (HMGCRi) had a positive correlation effect on the risk of breast cancer (OR: 0.8266, <i>p</i> = 0.0001, 95%CI: 0.7300–0.9232), ER+ breast cancer (OR: 0.8281, <i>p</i> = 0.0013, 95%CI: 0.7130–0.9431), serous ovarian cancer (OR: 0.7564, <i>p</i> = 0.0213, 95%CI: 0.5187–0.9941), overall ovarian cancer (OR: 0.5954, <i>p</i> = 2.25081E-05, 95%CI: 0.3555–0.8352), kidney cancer (OR: 0.9883, <i>p</i> = 5.85958E-11, 95% CI: 0.9848–0.9918), brain cancer (OR: 0.9982, <i>p</i> = 0.0168, 95%CI: 0.9967–0.9997) and overall ca","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 2","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-06-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70026","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144473049","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}
{"title":"Mitochondrial Ribosomal Protein Family in Cancers: Mechanistic Insights and Therapeutic Implications","authors":"Qian Chen, Yingli Zhang, Jin-Jian Lu, Ting Li","doi":"10.1002/mog2.70024","DOIUrl":"https://doi.org/10.1002/mog2.70024","url":null,"abstract":"<p>Mitochondria, as the main site for aerobic respiration in cells, are indispensable participants in the reprogrammed metabolic activities of tumor cells. Mitochondrial ribosomal proteins (MRPs), essential components of the mitochondrial ribosome (mitoribosome), play a critical role in maintaining mitochondrial function and regulating oncogenic signaling. Their molecular mechanisms and biological functions make MRPs key regulators of tumorigenesis, drug resistance, and tumor immune escape. MRPs are abnormally expressed in various cancer types and are linked to the prognosis of cancer patients. However, a thorough grasp of the specific mechanisms and a holistic analysis of the relationship between MRPs and different cancers are lacking. This review highlights the specific regulatory roles of MRPs, including MRPS5, MRPS29, MRPL9, MRPL12, MRPL13, MRPL33, MRPL58, and MRPL59, in cancer. Additionally, we examine the potential of MRPs as prospective clinical biomarkers and discuss their relationship with clinical prognosis and treatment response. We further explore the underlying reasons for the diverse functions of MRPs, their implications in cellular signaling and tumor immunity, and consider the prospects for developing MRP inhibitors as therapeutic strategies. Our comprehensive analysis deepens the understanding of MRPs complex biological functions and emphasizes their promising potential as therapeutic targets in cancer treatment.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 2","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-06-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70024","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144289297","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}
{"title":"Foe to Ally: Oncolytic Virus-Driven Xenorejection Ignites Potent Antitumor Immunity","authors":"Yanglin Xu, Bingcheng Chang, Wei He, Jia Liu","doi":"10.1002/mog2.70025","DOIUrl":"https://doi.org/10.1002/mog2.70025","url":null,"abstract":"<p>In a groundbreaking study published in the journal <i>Cell</i>, Yongxiang Zhao and collaborators designed a recombinant oncolytic virus (NDV-GT) by introducing the porcine α1,3-galactosyltransferase (<i>α1,3GT</i>) gene into the Newcastle disease virus (NDV). The engineered virus specifically infects tumor cells to express immunogenic αGal epitopes, thereby leveraging xenogeneic rejection mechanisms to activate robust antitumor immune responses (Figure 1) [<span>1</span>]. The innovative oncolytic virus demonstrates significant safety and preliminary efficacy in a cynomolgus monkey liver cancer model and a clinical trial enrolling 20 patients with refractory cancers.</p><p>Oncolytic viruses (OVs) have emerged as a promising and versatile therapeutic modality in cancer immunotherapy, owing to their ability for selective oncolysis and induction of systemic antitumor immunity [<span>2</span>]. However, inadequate immune priming, limited intra-tumoral dissemination and propagation, and rapid neutralization significantly hinder their therapeutic efficacy and clinical translation [<span>3</span>]. Moreover, the immunosuppressive tumor microenvironment (TME) markedly suppresses the outcomes of OV-based treatments. Despite advancements in engineered OVs and combinatorial regimens designed to enhance immune activation, clinical response rates remain unsatisfactory, indicating the urgent need for innovative strategies [<span>4</span>].</p><p>Xenogeneic rejection is a hyperacute immune response that occurs when heterologous organs or tissues are transplanted into a recipient of another species. The robust rejection could rapidly destroy xenografts by complement activation and endothelial damage [<span>5</span>]. The αGal epitope, a unique carbohydrate antigen, is widely present in non-primate mammals (e.g., pigs) and New World monkeys. However, during evolution, the ancestors of Old World monkeys and humans lost the αGal epitopes due to genetic mutations, but evolved anti-Gal antibodies to recognize the epitopes. Thus, when the organs or tissues from non-primate mammals are transplanted into humans, the anti-Gal antibodies quickly bind to αGal epitopes of xenografts, activating hyperacute rejection. Inspired by the hyperacute xenogeneic rejection, a porcine antigen was integrated into the Newcastle disease virus (NDV-GT) to deliver xenogeneic αGal epitopes to tumor cells (Figure 1A). Such recombinant virus camouflages porcine antigens on malignant cells, which would be recognized by the human body as heterologous transplant organs, thereby eliciting tumor-targeted hyperacute rejection analogous to xenograft destruction and converting the “cold” immunosuppressive TME to “hot” (Figure 1B).</p><p>The NDV-GT virus specifically and effectively infected different types of cancer cells <i>in vitro</i> as well as the parent virus, while exhibiting minimal infection in noncancerous cells. Moreover, the infected cancer cells effectively expressed the exogenous gen","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 2","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70025","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144213769","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}
{"title":"Activation of Adenosine Phosphate Signaling Promotes Antitumor Immunity in Tumor Microenvironment and Facilitate Immunotherapy","authors":"Yantao Xu, Ying Wang, Zixi Jiang, Yi He, Guowei Zhou, Benliang Wei, Jiachen Liu, Xiang Chen","doi":"10.1002/mog2.70022","DOIUrl":"https://doi.org/10.1002/mog2.70022","url":null,"abstract":"<p>Adenosine 5′-triphosphate (ATP) plays a crucial role in intracellular energetic metabolism and functions as a signal transducer in shaping the tumor microenvironment (TME). However, the understanding of the biological functions of adenosine phosphate signaling and its clinical relevance remains limited. Here, we deciphered the multi-omics dysregulation of 15 purinergic P2 receptors (P2Rs) and their clinical relevance. We revealed the presence of 5 ATP signaling subtypes in melanoma, with two distinct functional metaprograms—one metabolic and the other inflammatory. We developed an adenosine phosphate signaling model (APsig) that showed promising prognostic value in melanoma, as well as predictive efficacy of immunotherapy across 1068 tumor samples in 9 independent public cohorts. High APsig was associated with longer overall survival (OS) and improved response to tumor immunotherapy. Additionally, through single-cell and spatial transcriptomic analysis, we explored how APsig promotes antitumor immunity by activating myeloid lineage cells for antigen presentation. Our comprehensive characterization of P2R-mediated adenosine phosphate signaling at both bulk/single-cell and spatial transcriptomic levels highlights its potential as a promising target for developing novel anticancer agents, particularly in combination with immune checkpoint inhibitors.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 2","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-05-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70022","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144131579","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}
Xiao-Hong Ding, Xue-Pei Li, Fenfang Chen, Han Wang, Yi-Zhou Jiang
{"title":"Unlocking T-Cell Plasticity in the Tumor Microenvironment: Implications for Cancer Progression and Therapeutic Strategies","authors":"Xiao-Hong Ding, Xue-Pei Li, Fenfang Chen, Han Wang, Yi-Zhou Jiang","doi":"10.1002/mog2.70023","DOIUrl":"https://doi.org/10.1002/mog2.70023","url":null,"abstract":"<p>The tumor microenvironment (TME) is a complex and dynamic ecosystem crucial for cancer development and progression. Within this intricate milieu, T-cells constitute a heterogeneous population and serve as a cornerstone of antitumor immunity. Notably, T-cells can rapidly transition across a wide spectrum of phenotypic and functional states within the disrupted TME. Despite the crucial role of T-cells in cancer immunity, a comprehensive understanding of their plasticity within the TME remains limited. In this review, we delve into the functional plasticity and spatial distribution of T-cells in response to diverse microenvironmental conditions. Additionally, we review the plasticity of T-cell functional states during conventional therapies, highlighting their potential to enhance or limit therapeutic outcomes. Finally, we propose innovative therapeutic approaches that leverage T-cell plasticity to enhance clinical efficacy by regulating the immune response within the TME. By providing insights into the dynamics of T-cell behavior, this review highlights the promising potential of targeting T-cell plasticity as an immuno-sensitizer to refine therapeutic strategies and overcome current challenges in cancer treatment.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 2","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-05-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70023","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144131580","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}
{"title":"Oxidative Stress in Antigen Processing and Presentation","authors":"Qinxia Chang, Yaying Zhang, Xiaojun Liu, Peng Miao, Wenbing Pu, Shanshan Liu, Jing Zhang, Yuan Tian, Guobo Shen, Na Xie","doi":"10.1002/mog2.70020","DOIUrl":"https://doi.org/10.1002/mog2.70020","url":null,"abstract":"<p>Antigen processing and presentation are fundamental for connecting innate and adaptive immune responses in combating cancers and infections. Reactive oxygen species (ROS), serving as second messengers in various physiological processes, play a vital role in modulating antigen processing and presentation. However, oxidative stress due to an imbalance characterized by excessive accumulation of ROS or inadequate antioxidant defenses can severely impair antigen-specific immune responses, contributing to the pathophysiology of multiple health conditions, notably including various cancers, cancer-associated infections and autoimmune diseases. This review comprehensively investigates the multifaceted effects of ROS on antigen processing and presentation, encompassing immunopeptide generation, the functionality of antigen-presentation machinery (APM), and the interactions of antigen-presenting cells and antigen-specific effector cells. It emphasizes the critical pathophysiological roles of oxidative stress in diseases such as cancers, cancer-associated infections and autoimmune diseases. Moreover, we delve into the therapeutic potential of targeting redox homeostasis to enhance antitumor immune responses. By illuminating the intricate interplay between ROS and immune functionality, this review provides an essential theoretical framework for developing innovative immunotherapy strategies aimed at restoring immune competency and improving clinical outcomes in patients with immune-related diseases.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 2","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-05-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70020","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143950011","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}
Xingwen Wang, Minqiao Lu, Yi Zhang, Jiangwen Ma, Ying Hu
{"title":"Phase Separation: A New Dimension to Understanding Tumor Biology and Therapy","authors":"Xingwen Wang, Minqiao Lu, Yi Zhang, Jiangwen Ma, Ying Hu","doi":"10.1002/mog2.70018","DOIUrl":"https://doi.org/10.1002/mog2.70018","url":null,"abstract":"<p>Liquid–liquid phase separation (LLPS) plays a critical role in orchestrating various cellular processes, such as gene expression, signal transduction, and protein synthesis, by compartmentalizing cellular components without membrane boundaries. Emerging research has illuminated how dysregulated LLPS is integral to cancer development by influencing tumorigenesis, metastasis, immune system evasion, and resistance to therapy. The subtle differences in LLPS are crucial for understanding cancer progression and finding new treatments. However, despite its significant implications in oncology, the potential of specifically targeting LLPS in cancer therapy has not been thoroughly investigated. This review delves into the mechanisms of LLPS, exploring physiological triggers and their consequences in cancer biology. We discuss the profound impact of LLPS on the hallmarks of cancer and outline innovative strategies aimed at targeting LLPS. These strategies include the direct inhibition of phase condensate formation and the modulation of related signaling pathways. Although targeting LLPS poses several challenges, such as specificity and delivery methods, it represents a promising frontier in cancer treatment, potentially revolutionizing how we approach cancer therapy. This review emphasizes the academic and therapeutic importance of LLPS, advocating for it as an exciting and valuable target for future cancer treatment strategies.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 2","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-05-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70018","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143904963","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}
Jiaying Li, Mingshu Gao, Yanan Zhang, Dawen Liao, Feng Zhou, Zhaohui Zhang, Lele Ji, Yilin Zhao, Qichao Huang, Qian Bi, Nan Wang
{"title":"Alcohol Dehydrogenase 4-Mediated Retinol Metabolism Inhibits Hepatocellular Carcinoma Progression Through Inhibiting the Wnt/β-Catenin Pathway","authors":"Jiaying Li, Mingshu Gao, Yanan Zhang, Dawen Liao, Feng Zhou, Zhaohui Zhang, Lele Ji, Yilin Zhao, Qichao Huang, Qian Bi, Nan Wang","doi":"10.1002/mog2.70021","DOIUrl":"https://doi.org/10.1002/mog2.70021","url":null,"abstract":"<p>Hepatocellular carcinoma (HCC) ranks third in global cancer-related mortality, with limited therapies for advanced stages. Retinol, the alcohol form of vitamin A, has long been associated with liver diseases. Plasma retinol levels have been inversely correlated with the risk and poor prognosis of HCC. In this study, transcriptome data analysis identified retinol metabolism as the seventh KEGG-dysregulated pathway in cirrhosis tissue, ascending to the top position in HCC tissue compared to normal tissue. Specifically, a consistent downregulation of ADH4 (alcohol dehydrogenase 4), the retinol dehydrogenase among human ADHs, was observed, which correlated with poor prognosis in HCC patients. In vivo experiments demonstrated that silencing ADH4 enhances liver fibrosis and the progression of HCC. Mechanistically, ADH4 elevated intracellular levels of RA (retinoic acid), a biologically active derivative of retinol. RA-activated retinoid receptors RARs/RXRs, leading to inhibition of the downstream Wnt/β-catenin pathway and thereby hindering HCC progression. In contrast, the knockdown of ADH4 in hepatocytes triggers apoptosis. Notably, additional results demonstrated that the combined treatment of RA and cisplatin achieved synergistic antitumor effects in a mouse HCC model. In summary, our research elucidates that ADH4-mediated RA production suppresses HCC growth, providing a theoretical foundation for HCC treatment.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 2","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-04-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70021","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143871680","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}
{"title":"Fexofenadine Overcomes Osimertinib Resistance by Inhibiting c-Met in Non-Small Cell Lung Cancer","authors":"Kenneth K. W. To, Kwong-Sak Leung, William C. Cho","doi":"10.1002/mog2.70019","DOIUrl":"https://doi.org/10.1002/mog2.70019","url":null,"abstract":"<p>Osimertinib is the only third-generation EGFR tyrosine kinase inhibitor clinically approved for first-line treatment of advanced NSCLC patients harboring EGFR mutations. However, drug resistance severely hinders its clinical efficacy. Acquired <i>MET</i> amplification is an important mechanism causing osimertinib resistance. This study is the first to identify fexofenadine, originally indicated for allergic rhinitis and chronic urticaria, as a putative Met-inhibitor by in silico chemical-protein interactome analysis of known Met inhibitors. Fexofenadine was verified to inhibit recombinant Met kinase in cell-free assay and phosphorylation of Met and other downstream signaling molecules in osimertinib-resistant NSCLC cell lines. KINOME profiling revealed a similar kinase inhibition profile between fexofenadine and a known Met-inhibiting drug cabozantinib using Spearman rank-order correlation analysis. Among the tested osimertinib-resistant NSCLC cell lines, fexofenadine was the most efficacious in potentiating osimertinib in NCI-H820 (having <i>MET</i> amplification and EGFR-T790M mutation). Transcriptome profiling in NCI-H820 revealed that the differentially expressed genes following fexofenadine treatment were enriched in epithelial-mesenchymal transition-related biological pathways. Importantly, fexofenadine was also shown to significantly potentiate the antitumor effect of osimertinib in a drug-refractory NSCLC patient-derived tumor xenograft model in NSG mice, without inducing notable adverse effects. These findings advocate the clinical evaluation of repurposing fexofenadine to overcome osimertinib resistance.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 2","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-04-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70019","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143831113","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}