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}
{"title":"Characteristic, Regulation and Targeting Strategies of Cancer Stem Cells and Their Niche in Digestive System Tumors","authors":"Zhenzhen Chen, Huanle Qi, Yapeng Xue, Yaqi Zhang, Zhuo Zhang, Shun Xu, Shixin Liao, Xiaoyu Zeng, Jiayi Wu, Xinrui Lv, Qiankun He, Pingping Zhu","doi":"10.1002/mog2.70015","DOIUrl":"https://doi.org/10.1002/mog2.70015","url":null,"abstract":"<p>Digestive system tumor, including esophageal tumor, gastric tumor, intestinal tumor, liver tumor, pancreatic tumor, and cholangiocarcinoma, are the most common tumors worldwide and serve as a major cause of tumor-related death. Cancer stem cells (CSCs) are a small group of cells in tumors that harbor self-renewal, differentiation abilities, playing a crucial role in tumor initiation, progression, metastasis, and are supposed to be the fundamental cause of tumor recurrence after conventional treatment. A comprehensive understanding and targeting of CSCs is the key to overcoming tumors. In this review, focusing on digestive system tumors, we summarize the characteristics of CSCs, review the intracellular mechanisms that regulate self-renewal and functional maintenance of CSCs, including stemness pathways, transcription and epigenetic regulation, metabolic regulation, and noncoding RNAs, and demonstrate microenvironmental regulation and systemic regulation of CSCs at molecular and cellular levels. Finally, we summarize recent advances in tumor therapy with CSC targeting and their niche remodeling. These research progress on CSCs in digestive system tumors provide crucial insights into the occurrence, development, drug resistance, recurrence and metastasis of tumors, and offers new targeted treatment strategies for defeating tumors.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 2","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-04-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70015","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143801582","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":"Intercellular Mitochondrial Transfer Enhances the Antitumor Immunity of CD8+ T Cells","authors":"Ce Guo, Qiqing Yang, Long Zhang","doi":"10.1002/mog2.70017","DOIUrl":"https://doi.org/10.1002/mog2.70017","url":null,"abstract":"<p>In their paper published in <i>Cell</i> [<span>1</span>], Baldwin et al. used advanced techniques such as single-cell RNA sequencing, field emission scanning electron microscopy (FESEM), and confocal microscopy to systematically investigate the process by which bone marrow stromal cells (BMSCs) transfer mitochondria to CD8<sup>+</sup> T cells via tunneling nanotubes (TNTs). Through a series of experiments, they revealed how this process enhances T cell metabolic adaptability and antitumor efficacy, thus establishing mitochondrial transfer as an organelle transplantation strategy for significantly boosting T cell metabolic resilience and antitumor potential.</p><p>Adoptive T cell therapy (ACT) is a personalized immunotherapy; however, its efficacy against solid tumors is often limited because of the suppressive tumor microenvironment, which impairs T cell mitochondrial function, leading to T cell exhaustion and reduced antitumor immunity [<span>2</span>]. Recent research has demonstrated mitochondrial transfer across different cell types, which can repair damaged cells and in some cases, support tumor growth by providing mitochondria to tumor cells.</p><p>At present, tunneling nanotubes (TNTs) are recognized as a major pathway for mitochondrial transfer. These structures, supported by F-actin, span considerable distances between cells, facilitating the intercellular exchange of cytoplasmic materials and organelles [<span>3</span>]. However, whether mitochondrial transfer could restore mitochondrial function in exhausted T cells and present a new avenue for T cell–targeted solid tumor therapy remained unclear until Baldwin et al. provided crucial evidence supporting this model.</p><p>Within coculture systems, the researchers observed interactions between BMSCs and CD8<sup>+</sup> T cells, with field emission scanning electron microscopy (FESEM) capturing the formation of nanotubes between the two cell types. These nanotubes created intercellular “bridges” that enabled the transfer of mitochondria and other organelles from BMSCs to T cells. Confocal imaging analysis revealed a significant increase in mtDNA content within CD8<sup>+</sup> T cells (referred to as Mito<sup>+</sup> T cells) that had received mitochondria, confirming the occurrence of mitochondrial transfer from BMSCs. Further mechanistic investigation using gene enrichment analysis and immunoprecipitation sequencing revealed that Talin 2 (TLN2) acted as a key mediator of mitochondrial transfer via TNTs from BMSCs to CD8<sup>+</sup> T cells, highlighting its essential role in initiating nanotube formation in BMSCs and facilitating mitochondrial transfer (Figure 1).</p><p>To assess how mitochondrial transfer influences the metabolic performance of T cells, the researchers analyzed the oxygen consumption rate (OCR) of CD8<sup>+</sup> T cells, focusing on parameters such as basal respiration and spare respiratory capacity. The results indicated that Mito<sup>+</sup> T cells exhibited sign","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 2","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-04-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70017","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143778368","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":"RREB1: A Critical Transcription Factor, Integrates TGF-β and RAS Signals to Drive Cancer Metastasis via Regulation of Enhancers","authors":"Zhe Wang, Feng Xie, Fangfang Zhou","doi":"10.1002/mog2.70016","DOIUrl":"https://doi.org/10.1002/mog2.70016","url":null,"abstract":"<p>A recent research article published by Lee et al. [<span>1</span>] in <i>Cell</i> revealed that transforming growth factor β (TGF-β) and rat sarcoma viral oncogene homolog (RAS) signaling, together trigger expression of epithelial-to-mesenchymal transition (EMT) and fibrogenic factors enhancing cancer metastasis through a precise and complex system. The authors elucidated that RAS-responsive element-binding protein 1 (RREB1)-mediated TGF-β-dependent fibrogenesis, and EMT come together to form a program to regulate cancer metastasis (Figure 1). This study enhances our understanding of the crosstalk between RAS and TGF-β in cancer metastasis, providing a potential therapeutic target.</p><p>RREB1, comprising 15 zinc finger (ZF) domains, is a critical transcription factor downstream of the RAS/mitogen-activated protein kinase (MAPK) signaling cascade, which plays a significant role in integration of RAS and TGF-β signaling pathways. TGF-β-activated small mother against decapentaplegic (SMAD) transcription factors are recruited by MAPK-activated RREB1 to Snail family transcriptional repressor (SNAIL). The recruitment of SMADs to SNAIL increases expression of SNAIL and triggers induction of developmental and fibrogenic EMT in carcinoma cells [<span>2, 3</span>]. Furthermore, RREB1-eukaryotic translation elongation factor 1A1 (eEF1A1)-3′ UTR axis enhances the translation of mitochondrial respiratory complex proteins encoded in nucleus and offers a novel therapeutic target for combating leukemia stem cells (LSCs) [<span>4</span>].</p><p>Cancer metastasis is the primary cause of patient mortality. During cancer metastasis, EMT is a crucial process in which epithelial cells lose their typical characteristics and acquire traits of mesenchymal cells, enhancing cell migration, invasion of surrounding tissues, and resistance to treatments. Su et al. [<span>2</span>] and Fontana et al. [<span>5</span>] revealed that the synergy between the TGF-β and RAS pathways trigger the EMT in fibrogenesis. Additionally, they identified RREB1, a RAS transcriptional effector, as an important cofactor of SMAD inducing EMT-transcription factors' (TFs) expression. Furthermore, in human acute myeloid leukemia (AML), a short variant of RREB1, known as RREB1S (1368 bp), enhances translation of nuclear-encoded mitochondrial genes mediated through its interaction with the translational factor eEF1A1, to maintain the characteristics of LSCs [<span>4</span>]. However, the subset of TGF-β mediated EMT-TFs regulated by RREB1 and the specific mechanism of RREB1-integrated RAS and TGF-β signaling transduction in cancer metastasis remains unknown.</p><p>Activated mutant-Kirsten RAS (KRAS) is a key driver mutation in lung adenocarcinoma (LUAD) accounting for one of the most common genetic subsets of human LUAD [<span>1</span>]. Single cell RNA-sequencing and immunofluorescence findings of metastasis samples isolated from <i>KRAS</i>-mutated patients with LUAD suggested that EMT-TFs and f","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-03-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70016","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143629856","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}
Hanchuan Shen, Bing Liu, Hangyu Zhang, Yang Liu, Chenggang Li
{"title":"Adjuvant Therapy Combining Donafenib and Sintilimab Enhances Recurrence-Free Survival in Hepatocellular Carcinoma Patients With High-Risk Recurrence Factors After Radical Resection: A Retrospective Cohort Study","authors":"Hanchuan Shen, Bing Liu, Hangyu Zhang, Yang Liu, Chenggang Li","doi":"10.1002/mog2.70013","DOIUrl":"https://doi.org/10.1002/mog2.70013","url":null,"abstract":"<p>Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer death and the sixth in incidence globally. Curative resection is a main treatment for HCC patients, yet the 5-year post-radical resection tumor recurrence rate remains up to 70%. Most high-risk recurrence patients may experience early recurrence within 2 years and have a poor prognosis [<span>1</span>]. Notably, there's still a lack of standardized postoperative treatment globally. A recent study, IMbrave050, found that the postoperative adjuvant use of the combination of bevacizumab and atezolizumab did not extend recurrence-free survival (RFS) when compared to surveillance [<span>2</span>]. Combinations of tyrosine kinase inhibitors (TKIs) and PD-1 inhibitors have shown promise in unresectable HCC, with the efficacy of adjuvant treatment postradical resection yet to be confirmed. Donafenib, a novel TKI, has demonstrated survival benefits over sorafenib in unresectable HCC. Additionally, several studies of donafenib combined with PD-1 inhibitors have shown promising results for unresectable HCC because of synergistic antitumor effects [<span>3, 4</span>]. This retrospective study aims to evaluate the efficacy and safety of adjuvant donafenib combined with PD-1inhibitor sintilimab in preventing tumor recurrence in HCC patients with high-risk recurrence factors (HRRF).</p><p>This retrospective study involved patients who underwent radical resection from June 2019 to February 2023. Informed consent was waived due to the retrospective nature of the study. The study adhered to the Helsinki Declaration and was approved by the ethics committee of Chinese PLA General Hospital (Approval no. S2023-737-01). Eligible patients were ≥ 18 years old and had HRRF, including tumor size ≥ 5 cm, tumor number ≥ 2, macrovascular invasion (invasion of portal and hepatic veins), presence of microvascular invasion (MVI), and satellite nodules. Patients receiving adjuvant donafenib (0.1 g twice daily orally) and sintilimab (200 mg every 3 weeks intravenously) initiated treatment 4–8 weeks postsurgery for up to 1 year or until HCC recurrence or serious adverse events. To balance confounding factors, we utilized a 1:1 propensity score matching (PSM) analysis with a caliper width of 0.02, which considered variables such as sex, age, Eastern Co-operative Oncology Group Performance Status, Barcelona Clinic Liver Cancer (BCLC) stage, Child-Pugh score, HBV, tumor diameter, tumor number, macrovascular invasion, MVI, satellite nodules, alpha-fetoprotein (AFP), and tumor differentiation. Imaging was conducted every 12 weeks during the first 2 years and then every 24 weeks until disease recurrence. The outcomes included RFS, overall survival (OS) and safety.</p><p>From June 2019 to February 2023, we collected 260 HCC patients with HRRF after radical resection. A total of 101 patients met the inclusion criteria and were ultimately included in the study. The treatment group consisted of 34 patients who received a","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-03-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70013","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143554596","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":"Macrophage-Mediated Myelin Recycling Promotes Malignant Development of Glioblastoma","authors":"Huanhuan Wang, Long Zhang, Feng Xie","doi":"10.1002/mog2.70014","DOIUrl":"https://doi.org/10.1002/mog2.70014","url":null,"abstract":"<p>A recent study published in the journal <i>Cell</i> [<span>1</span>] revealed the critical role of macrophages in the malignant development of brain cancers, particularly glioblastoma. Through a series of in-vivo and in-vitro experiments, the study researched how macrophages provide essential lipids and nutrients to brain cancer cells by mediating the recycling and reuse of myelin, the insulating layer in the nervous system, which in turn promotes growth and invasion of tumor. They explored the heterogeneity of tumor-associated macrophages (TAM) in glioblastoma tumor microenvironment using single-cell and multi-omics analyses and revealed their specific interactions with different glioblastoma subtypes. The study reported the dynamic contexture of the glioblastoma tumor microenvironment at single-cell levels during primary or recurrent tumor progression, revealed the colocated diversity of niche-specific interactions between TAMs and glioblastoma subtypes at spatial transcriptomic levels, evaluated the chromatin landscape changes and immuno-suppressive features associated with the lipid-laden phenotype using multi-omics sequences, addressed the transfer route of lipid flux from myelin to macrophages at last to mesenchymal-like (MES-like) glioblastoma cells, demonstrated the intrinsic lipid traffic in macrophages and the altered metabolic manner in glioblastoma cells using lipidomics analysis and experiments, presented the protumorigenic functions of lipid-laden macrophages (LLMs) in glioblastoma and their relevance to clinical survival or immunotherapeutic response.</p><p>The study found that in the brain tumor microenvironment, macrophages are able to take up and accumulate myelin debris in large quantities. These myelin fragments are converted by macrophages into cholesterol and other lipids, which are then delivered to brain cancer cells to support their growth and malignant transformation. They also found that specific types of macrophages, such as TAMs with high glycoprotein nonmetastatic melanoma protein B (GPNMB) expression, are closely associated with areas of high myelin debris accumulation and exhibit unique patterns of lipid metabolism and inflammatory activity. The study further demonstrated that macrophage-mediated lipid delivery not only provides an energy source for brain cancer cells, but also promotes the invasion and metastasis ability of cancer cells. By interfering with the lipid metabolism pathway of macrophages, the progress of brain cancer can be significantly inhibited. When macrophages take in myelin fragments, their inflammatory activity is suppressed and they shift to an “anti-inflammatory” state. This anti-inflammatory state may help maintain the stability of the tumor microenvironment, thus providing favorable conditions for tumor cell growth. The study was also verified using patient sample data and found similar patterns of macrophage activity in the tumor microenvironment of glioma patients, which suggests that ","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-02-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70014","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143481329","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}
Lin Tang, Shao-Cong Peng, Xiao-Wan Zhuang, Yan He, Yu-Xiang Song, Hao Nie, Can-Can Zheng, Zhen-Yu Pan, Alfred King-Yin Lam, Ming-Liang He, Xing-Yuan Shi, Bin Li, Wen Wen Xu
{"title":"Tumor Metastasis: Mechanistic Insights and Therapeutic Intervention","authors":"Lin Tang, Shao-Cong Peng, Xiao-Wan Zhuang, Yan He, Yu-Xiang Song, Hao Nie, Can-Can Zheng, Zhen-Yu Pan, Alfred King-Yin Lam, Ming-Liang He, Xing-Yuan Shi, Bin Li, Wen Wen Xu","doi":"10.1002/mog2.70012","DOIUrl":"https://doi.org/10.1002/mog2.70012","url":null,"abstract":"<p>Metastasis remains a leading cause of cancer-related deaths, defined by a complex, multi-step process in which tumor cells spread and form secondary growths in distant tissues. Despite substantial progress in understanding metastasis, the molecular mechanisms driving this process and the development of effective therapies remain incompletely understood. Elucidating the molecular pathways governing metastasis is essential for the discovery of innovative therapeutic targets. The rapid advancements in sequencing technologies and the expansion of biological databases have significantly deepened our understanding of the molecular drivers of metastasis and associated drug resistance. This review focuses on the molecular drivers of metastasis, particularly the roles of genetic mutations, epigenetic changes, and post-translational modifications in metastasis progression. We also examine how the tumor microenvironment influences metastatic behavior and explore emerging therapeutic strategies, including targeted therapies and immunotherapies. Finally, we discuss future research directions, stressing the importance of novel treatment approaches and personalized strategies to overcome metastasis and improve patient outcomes. By integrating contemporary insights into the molecular basis of metastasis and therapeutic innovation, this review provides a comprehensive framework to guide future research and clinical advancements in metastatic cancer.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-02-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70012","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143423918","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}
Xiaohui Du, Congcong Zhang, Ying Li, Peipei He, Jian Wang, Xuena Chen, Han Wang, Qi Wang
{"title":"Lomitapide: Targeting METTL3 to Overcome Osimertinib Resistance in NSCLC Through Autophagy Activation","authors":"Xiaohui Du, Congcong Zhang, Ying Li, Peipei He, Jian Wang, Xuena Chen, Han Wang, Qi Wang","doi":"10.1002/mog2.70011","DOIUrl":"https://doi.org/10.1002/mog2.70011","url":null,"abstract":"<p>Osimertinib resistance remains a significant challenge in the treatment of non-small cell lung cancer (NSCLC). <i>N</i><sup>6</sup>-methyladenosine (m<sup>6</sup>A) modifications are closely linked to various mechanisms of anticancer resistance and autophagy, offering new avenues for targeted therapies. However, the role of m<sup>6</sup>A-mediated autophagy in osimertinib-resistant NSCLC is still unclear. In this study, we utilized multi-omics sequencing analysis and found that overexpression of the m<sup>6</sup>A methyltransferase METTL3 contributes to osimertinib resistance in NSCLC. Importantly, we identified that METTL3 positively regulates the expression of the autophagy-related gene ubiquinone-cytochrome C reductase complex assembly factor 2 (<i>UQCC2</i>) through an m<sup>6</sup>A-dependent mechanism. Further, we confirmed that <i>METTL3</i> knockdown leads to <i>UQCC2</i> downregulation and triggers autophagy activation. Interestingly, lomitapide, a cholesterol-lowering drug, was repurposed to enhance the sensitivity of cancer cells to therapy by inhibiting METTL3, which in turn activated autophagy-associated cell death pathways, reversing osimertinib resistance. This study emphasizes the critical role of the METTL3/UQCC2 axis in autophagy-mediated drug resistance and positions lomitapide as a promising METTL3 inhibitor and autophagy inducer with potential therapeutic effects, either alone or in combination with other anticancer agents, in patients with osimertinib-resistant NSCLC.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-01-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70011","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143120497","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}
Nan Liu, Wenwen Wei, Kexing Ren, Dandan Liang, Dong Yang, Weishan Zhang, Beibei Yang, Bin Sun, Jincheng Zhao, Dan Cao, Liqun Zou, Xudong Zhao
{"title":"Ephrin A1 ligand-based CAR-T cells for immunotherapy of EphA2-positive cancer","authors":"Nan Liu, Wenwen Wei, Kexing Ren, Dandan Liang, Dong Yang, Weishan Zhang, Beibei Yang, Bin Sun, Jincheng Zhao, Dan Cao, Liqun Zou, Xudong Zhao","doi":"10.1002/mog2.70010","DOIUrl":"https://doi.org/10.1002/mog2.70010","url":null,"abstract":"<p>Chimeric antigen receptor (CAR) T cells have demonstrated promising results in hematological malignancies; however, challenges remain in treating solid tumors. New CARs with more effectiveness and lower side effects are needed. Ephrin type-A receptor 2 (EphA2) belongs to the Ephrin family of receptor tyrosine kinases, which is overexpressed in several solid malignancies. Compared with some single-chain variable fragment (ScFv) CARs that exhibit excessively high affinity for their targets, natural receptor/ligand-based CARs maintain inherent affinity for their binding partners, potentially balancing cytotoxicity and side effects to better meet clinical needs. Here, we designed a CAR targeting EphA2-positive cancer cells by exploiting the extracellular domain of its natural ligand Ephrin A1 (EFNA1). EFNA1 CAR-T cells exhibited specific cytotoxicity against various cancer cells and cancer stem-like cells in vitro, and significantly suppressed tumor growth in a pancreatic cancer xenograft mouse model. Moreover, although these CAR-T cells specifically targeted mouse EphA2 and killed mouse tumor cell lines in vitro, they did not induce obvious side effects in mice. Additionally, it also showed good safety in <i>rhesus macaques</i>. Collectively, these results validate the therapeutic effectiveness and safety of EFNA1 CAR-T cells for treating solid tumors.</p>","PeriodicalId":100902,"journal":{"name":"MedComm – Oncology","volume":"4 1","pages":""},"PeriodicalIF":0.0,"publicationDate":"2025-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mog2.70010","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143118772","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}