MedCommPub Date : 2024-11-20DOI: 10.1002/mco2.70012
Yusheng Zhang, Hong Wang, Yiwei Sang, Mei Liu, Qing Wang, Hongjun Yang, Xianyu Li
{"title":"Gut microbiota in health and disease: advances and future prospects","authors":"Yusheng Zhang, Hong Wang, Yiwei Sang, Mei Liu, Qing Wang, Hongjun Yang, Xianyu Li","doi":"10.1002/mco2.70012","DOIUrl":"https://doi.org/10.1002/mco2.70012","url":null,"abstract":"<p>The gut microbiota plays a critical role in maintaining human health, influencing a wide range of physiological processes, including immune regulation, metabolism, and neurological function. Recent studies have shown that imbalances in gut microbiota composition can contribute to the onset and progression of various diseases, such as metabolic disorders (e.g., obesity and diabetes) and neurodegenerative conditions (e.g., Alzheimer's and Parkinson's). These conditions are often accompanied by chronic inflammation and dysregulated immune responses, which are closely linked to specific forms of cell death, including pyroptosis and ferroptosis. Pathogenic bacteria in the gut can trigger these cell death pathways through toxin release, while probiotics have been found to mitigate these effects by modulating immune responses. Despite these insights, the precise mechanisms through which the gut microbiota influences these diseases remain insufficiently understood. This review consolidates recent findings on the impact of gut microbiota in these immune-mediated and inflammation-associated conditions. It also identifies gaps in current research and explores the potential of advanced technologies, such as organ-on-chip models and the microbiome–gut–organ axis, for deepening our understanding. Emerging tools, including single-bacterium omics and spatial metabolomics, are discussed for their promise in elucidating the microbiota's role in disease development.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"5 12","pages":""},"PeriodicalIF":10.7,"publicationDate":"2024-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mco2.70012","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142674221","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}
MedCommPub Date : 2024-11-20DOI: 10.1002/mco2.70010
Qian Zhou, Yu Meng, Jiayuan Le, Yuming Sun, Yating Dian, Lei Yao, Yixiao Xiong, Furong Zeng, Xiang Chen, Guangtong Deng
{"title":"Ferroptosis: mechanisms and therapeutic targets","authors":"Qian Zhou, Yu Meng, Jiayuan Le, Yuming Sun, Yating Dian, Lei Yao, Yixiao Xiong, Furong Zeng, Xiang Chen, Guangtong Deng","doi":"10.1002/mco2.70010","DOIUrl":"https://doi.org/10.1002/mco2.70010","url":null,"abstract":"<p>Ferroptosis is a nonapoptotic form of cell death characterized by iron-dependent lipid peroxidation in membrane phospholipids. Since its identification in 2012, extensive research has unveiled its involvement in the pathophysiology of numerous diseases, including cancers, neurodegenerative disorders, organ injuries, infectious diseases, autoimmune conditions, metabolic disorders, and skin diseases. Oxidizable lipids, overload iron, and compromised antioxidant systems are known as critical prerequisites for driving overwhelming lipid peroxidation, ultimately leading to plasma membrane rupture and ferroptotic cell death. However, the precise regulatory networks governing ferroptosis and ferroptosis-targeted therapy in these diseases remain largely undefined, hindering the development of pharmacological agonists and antagonists. In this review, we first elucidate core mechanisms of ferroptosis and summarize its epigenetic modifications (e.g., histone modifications, DNA methylation, noncoding RNAs, and N6-methyladenosine modification) and nonepigenetic modifications (e.g., genetic mutations, transcriptional regulation, and posttranslational modifications). We then discuss the association between ferroptosis and disease pathogenesis and explore therapeutic approaches for targeting ferroptosis. We also introduce potential clinical monitoring strategies for ferroptosis. Finally, we put forward several unresolved issues in which progress is needed to better understand ferroptosis. We hope this review will offer promise for the clinical application of ferroptosis-targeted therapies in the context of human health and disease.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"5 12","pages":""},"PeriodicalIF":10.7,"publicationDate":"2024-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mco2.70010","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142674278","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}
MedCommPub Date : 2024-11-19DOI: 10.1002/mco2.70000
Gemma Garcia-Vicién, Núria Ruiz, Patrick Micke, José Carlos Ruffinelli, Kristel Mils, María Bañuls, Natalia Molina, Miguel A. Pardo, Laura Lladó, Artur Mezheyeuski, David G. Molleví
{"title":"The histological growth patterns in liver metastases from colorectal cancer display differences in lymphoid, myeloid, and mesenchymal cells","authors":"Gemma Garcia-Vicién, Núria Ruiz, Patrick Micke, José Carlos Ruffinelli, Kristel Mils, María Bañuls, Natalia Molina, Miguel A. Pardo, Laura Lladó, Artur Mezheyeuski, David G. Molleví","doi":"10.1002/mco2.70000","DOIUrl":"https://doi.org/10.1002/mco2.70000","url":null,"abstract":"<p>Colorectal liver metastases grow following different histologic growth patterns (HGPs), classified as desmoplastic and nondesmoplastic (dHGP, non-dHGP), being the latter associated with worst prognosis. This study aimed to investigate the tumor microenvironment (TME) between HGPs supporting different survival. Multiplexed immunohistochemical staining was performed with the Opal7 system in a 100-patients cohort to evaluate the tumor–liver interface with three different cell panels: lymphoid, myeloid, and carcinoma-associated fibroblasts. Differences between HGPs were assessed by Mann–Whitney <i>U</i> test with Pratt correction and Holm–Bonferroni multitest adjustment. Cytotoxic T-cells were more abundant in tumoral areas of dHGP, while non-dHGP had higher macrophages infiltration, Th2, CD163<sup>+</sup>, and Calprotectin<sup>+</sup> cells as well as higher pSMAD2 expression. Regarding carcinoma-associated fibroblasts, several subsets expressing COL1A1 were enriched in dHGP, while αSMA<sup>low</sup>_single cells were present at higher densities in non-dHGP. Interestingly, Calprotectin<sup>+</sup> cells confer better prognoses in non-dHGP, identifying a subgroup of good outcome patients that unexpectedly also show an enrichment in other myeloid cells. In summary, our results illustrate different TME landscapes with respect to HGPs. dHGP presents a higher degree of immunocompetence, higher amounts of Collagen 1 as well as lesser presence of myeloid cell populations, features that might be influencing on the better prognosis of encapsulated metastases.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"5 12","pages":""},"PeriodicalIF":10.7,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mco2.70000","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142674369","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}
MedCommPub Date : 2024-11-14DOI: 10.1002/mco2.70011
Qingjian Chen, Yu-Hong Xu, Shiyang Kang, WuHao Lin, Linna Luo, Luping Yang, Qi-Hua Zhang, Pan Yang, Jia-Qian Huang, Xiaoni Zhang, Jing Zhang, Qi Zhao, Rui-Hua Xu, Hui-Yan Luo
{"title":"The tissue and circulating cell-free DNA-derived genetic landscape of premalignant colorectal lesions and its application for early diagnosis of colorectal cancer","authors":"Qingjian Chen, Yu-Hong Xu, Shiyang Kang, WuHao Lin, Linna Luo, Luping Yang, Qi-Hua Zhang, Pan Yang, Jia-Qian Huang, Xiaoni Zhang, Jing Zhang, Qi Zhao, Rui-Hua Xu, Hui-Yan Luo","doi":"10.1002/mco2.70011","DOIUrl":"https://doi.org/10.1002/mco2.70011","url":null,"abstract":"<p>Colorectal adenomas (CRAs) represent precancerous lesions that precede the development of colorectal cancer (CRC). Regular monitoring of CRAs can hinder the progression into carcinoma. To explore the utility of tissue DNA and circulating cell-free DNA (cfDNA) in early diagnosis of CRC, we retrospectively sequenced paired tissue and plasma samples from 85 patients with conventional CRAs. The genetic alterations identified were compared with those from 78 stage-I CRC patients (CRC-I) in the ChangKang project. Within the CRA cohort, we pinpointed 12 genes, notably <i>APC</i>, <i>KRAS</i>, and <i>SOX9</i>, that exhibited significant mutated rates in tissue. Patients harboring <i>KMT2C</i> and <i>KMT2D</i> mutations displayed persistent polyps. By comparing with the mutational profiles of metastatic CRC plasma samples, we found that <i>ZNF717</i> was exclusively mutated in CRAs, while <i>KMT2C</i> and <i>KMT2D</i> mutations were detected in both CRA and CRC. The presence of cfDNA mutations in plasma was validated through polymerase chain reaction, enhancing the feasibility of using cfDNA mutations for early CRC screening. Compared with CRC-I, CRAs exhibited a reduced frequency of <i>TP53</i> and <i>PIK3CA</i> somatic mutations and underwent non-neutral evolution more often. We established a random forest model based on 15 characteristic genes to distinguish CRA and CRC, achieving an area under the curve of 0.89. Through this endeavor, we identified two novel genes, <i>CNTNAP5</i> and <i>GATA6</i>, implicated in CRC carcinogenesis. Overall, our findings reveal convincing biomarkers markers for detecting CRAs with a propensity for CRC development, highlighting the importance of early genetic screening in CRC prevention.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"5 12","pages":""},"PeriodicalIF":10.7,"publicationDate":"2024-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/mco2.70011","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142642012","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":"LINC01764 promotes colorectal cancer cells proliferation, metastasis, and 5-fluorouracil resistance by regulating glucose and glutamine metabolism via promoting c-MYC translation","authors":"Ran Duan, Yujia Zhai, Qiushuang Wang, Liqin Zhao, Yixuan Wang, Nuoya Yu, Jieyun Zhang, Weijian Guo","doi":"10.1002/mco2.70003","DOIUrl":"10.1002/mco2.70003","url":null,"abstract":"<p>Few biomarkers are available for predicting chemotherapeutic response and prognosis in colorectal cancer (CRC). Long-noncoding RNAs (lncRNAs) are essential for CRC development and growth. Therefore, studying lncRNAs may reveal potential predictors of chemotherapy response and prognosis in CRC. LINC01764 was analyzed using datasets from Fudan University Shanghai Cancer Center's advanced CRC patients’ RNA-seq and The Cancer Genome Atlas datasets. Gene set enrichment analysis was employed to detect related pathways. Cotransfection experiments, RNA pulldown assays, RNA-binding protein immunoprecipitation, protein synthesis activity, and dual-luciferase reporter assays were performed to determine interactions among LINC01764, hnRNPK, and c-MYC. High LINC01764 expression correlates with metastasis, a poor response to FOLFOX/XELOX chemotherapy, and a poor prognosis in CRC. LINC01764 enhance glycolysis and glutamine metabolism to promote CRC cells proliferation, metastasis, and 5-fluorouracil (5-FU) resistance. LINC01764 specifically binds to hnRNPK, facilitating its interaction with c-MYC mRNA and promoting internal ribosome entry site (IRES)-dependent translation of c-MYC, thereby exerting oncogenic effects. LINC01764 induced 5-FU chemoresistance by upregulating the c-MYC, glucose, and glutamine metabolism pathways, which downregulated <i>UPP1</i>, crucial for activating 5-FU. Conclusively, LINC01764 promotes CRC progression and 5-FU resistance through hnRNPK-mediated-c-MYC IRES-dependent translational regulation, which suggests its potential as a predictor of CRC chemotherapy response and prognosis.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"5 11","pages":""},"PeriodicalIF":10.7,"publicationDate":"2024-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11555016/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142634079","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}
Shanshan Pan, Luan Yin, Jie Liu, Jie Tong, Zichuan Wang, Jiahui Zhao, Xuesong Liu, Yong Chen, Jing Miao, Yuan Zhou, Su Zeng, Tengfei Xu
{"title":"Metabolomics-driven approaches for identifying therapeutic targets in drug discovery","authors":"Shanshan Pan, Luan Yin, Jie Liu, Jie Tong, Zichuan Wang, Jiahui Zhao, Xuesong Liu, Yong Chen, Jing Miao, Yuan Zhou, Su Zeng, Tengfei Xu","doi":"10.1002/mco2.792","DOIUrl":"10.1002/mco2.792","url":null,"abstract":"<p>Identification of therapeutic targets can directly elucidate the mechanism and effect of drug therapy, which is a central step in drug development. The disconnect between protein targets and phenotypes under complex mechanisms hampers comprehensive target understanding. Metabolomics, as a systems biology tool that captures phenotypic changes induced by exogenous compounds, has emerged as a valuable approach for target identification. A comprehensive overview was provided in this review to illustrate the principles and advantages of metabolomics, delving into the application of metabolomics in target identification. This review outlines various metabolomics-based methods, such as dose–response metabolomics, stable isotope-resolved metabolomics, and multiomics, which identify key enzymes and metabolic pathways affected by exogenous substances through dose-dependent metabolite–drug interactions. Emerging techniques, including single-cell metabolomics, artificial intelligence, and mass spectrometry imaging, are also explored for their potential to enhance target discovery. The review emphasizes metabolomics' critical role in advancing our understanding of disease mechanisms and accelerating targeted drug development, while acknowledging current challenges in the field.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"5 11","pages":""},"PeriodicalIF":10.7,"publicationDate":"2024-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11555024/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142636163","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}
Mazaher Maghsoudloo, Khatere Mokhtari, Behdokht Jamali, Amir Gholamzad, Maliheh Entezari, Mehrdad Hashemi, Junjiang Fu
{"title":"Multifaceted role of TRIM28 in health and disease","authors":"Mazaher Maghsoudloo, Khatere Mokhtari, Behdokht Jamali, Amir Gholamzad, Maliheh Entezari, Mehrdad Hashemi, Junjiang Fu","doi":"10.1002/mco2.790","DOIUrl":"10.1002/mco2.790","url":null,"abstract":"<p>The <i>TRIM</i> (tripartite motif) family, with <i>TRIM28</i> as a key member, plays a vital role in regulating health and disease. <i>TRIM28</i> contains various functional domains essential for transcriptional regulation, primarily through its interaction with <i>KRAB-ZNF</i> proteins, which influence chromatin remodeling and gene expression. Despite extensive research, the precise mechanisms by which <i>TRIM28</i> impacts health and disease remain elusive. This review delves into <i>TRIM28</i>’s multifaceted roles in maintaining health, contributing to a variety of diseases, and influencing cancer progression. In cancers, <i>TRIM28</i> exhibits a dual nature, functioning as both a tumor promoter and suppressor depending on the cellular context and cancer type. The review also explores its critical involvement in processes such as DNA repair, cell cycle regulation, epithelial-to-mesenchymal transition, and the maintenance of stem cell properties. By uncovering <i>TRIM28</i>’s complex roles across different cancers and other diseases, this review underscores its potential as a therapeutic target. The significance of <i>TRIM28</i> as a versatile regulator opens the door to innovative therapeutic strategies, particularly in cancer treatment and the management of other diseases. Ongoing research into <i>TRIM28</i> may yield key insights into disease progression and novel treatment options.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"5 11","pages":""},"PeriodicalIF":10.7,"publicationDate":"2024-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11554878/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142634198","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":"Full-active pharmaceutical ingredient nanosensitizer for augmented photoimmunotherapy by synergistic mitochondria targeting and immunogenic death inducing","authors":"Xianghui Li, Haoran Wang, Zhiyan Li, Song Liu, Yuanyuan Chen, Zhuren Ruan, Zhijian Yao, Gao Wei, Cunwei Cao, Wenjun Zheng, Wenxian Guan","doi":"10.1002/mco2.756","DOIUrl":"10.1002/mco2.756","url":null,"abstract":"<p>The precise and effective activation of the immune response is crucial in promising therapy curing cancer. Photoimmunotherapy (PIT) is an emerging strategy for precise regulation and highly spatiotemporal selectivity. However, this approach faces a significant challenge due to the off-target effect and the immunosuppressive microenvironment. To address this challenge, a nanoscale full-active pharmaceutical ingredient (API) photo-immune stimulator was developed. This formulation overcomes the limitations of PIT by strengthening the ability to penetrate tumors deeply and inducing precise and potent mitochondria-targeted dual-mode photodynamic therapy and photothermal therapy. Along with inhibiting overexpressed Hsp90, this nanosensitizer in turn improves the immunosuppressive microenvironment. Ultimately, this mitochondria-targeted PIT demonstrated potent antitumor efficacy, achieving a remarkable inhibition rate of ≥95% for both established primary tumors and distant abscopal tumors. In conclusion, this novel self-delivery full-API nanosystem enhances the efficacy of phototherapy and reprograms the immunosuppressive microenvironment, thereby holding great promise in the development of precise and effective immunotherapy.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"5 11","pages":""},"PeriodicalIF":10.7,"publicationDate":"2024-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11550090/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142633924","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":"Synthetic circRNA therapeutics: innovations, strategies, and future horizons","authors":"Jingsheng Cai, Zonghao Qiu, William Chi-Shing Cho, Zheng Liu, Shaoyi Chen, Haoran Li, Kezhong Chen, Yun Li, Chijian Zuo, Mantang Qiu","doi":"10.1002/mco2.720","DOIUrl":"10.1002/mco2.720","url":null,"abstract":"<p>Small molecule drugs are increasingly emerging as innovative and effective treatments for various diseases, with mRNA therapeutics being a notable representative. The success of COVID-19 vaccines has underscored the transformative potential of mRNA in RNA therapeutics. Within the RNA family, there is another unique type known as circRNA. This single-stranded closed-loop RNA molecule offers notable advantages over mRNA, including enhanced stability and prolonged protein expression, which may significantly impact therapeutic strategies. Furthermore, circRNA plays a pivotal role in the pathogenesis of various diseases, such as cancers, autoimmune disorders, and cardiovascular diseases, making it a promising clinical intervention target. Despite these benefits, the application of circRNA in clinical settings remains underexplored. This review provides a comprehensive overview of the current state of synthetic circRNA therapeutics, focusing on its synthesis, optimization, delivery, and diverse applications. It also addresses the challenges impeding the advancement of circRNA therapeutics from bench to bedside. By summarizing these aspects, the review aims to equip researchers with insights into the ongoing developments and future directions in circRNA therapeutics. Highlighting both the progress and the existing gaps in circRNA research, this review offers valuable perspectives for advancing the field and guiding future investigations.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"5 11","pages":""},"PeriodicalIF":10.7,"publicationDate":"2024-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11550093/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142634692","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":"Circulating tumor DNA methylation detection as biomarker and its application in tumor liquid biopsy: advances and challenges","authors":"Lingyu Li, Yingli Sun","doi":"10.1002/mco2.766","DOIUrl":"10.1002/mco2.766","url":null,"abstract":"<p>Circulating tumor DNA (ctDNA) methylation, an innovative liquid biopsy biomarker, has emerged as a promising tool in early cancer diagnosis, monitoring, and prognosis prediction. As a noninvasive approach, liquid biopsy overcomes the limitations of traditional tissue biopsy. Among various biomarkers, ctDNA methylation has garnered significant attention due to its high specificity and early detection capability across diverse cancer types. Despite its immense potential, the clinical application of ctDNA methylation faces substantial challenges pertaining to sensitivity, specificity, and standardization. In this review, we begin by introducing the basic biology and common detection techniques of ctDNA methylation. We then explore recent advancements and the challenges faced in the clinical application of ctDNA methylation in liquid biopsies. This includes progress in early screening and diagnosis, identification of clinical molecular subtypes, monitoring of recurrence and minimal residual disease (MRD), prediction of treatment response and prognosis, assessment of tumor burden, and determination of tissue origin. Finally, we discuss the future perspectives and challenges of ctDNA methylation detection in clinical applications. This comprehensive overview underscores the vital role of ctDNA methylation in enhancing cancer diagnostic accuracy, personalizing treatments, and effectively monitoring disease progression, providing valuable insights for future research and clinical practice.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":"5 11","pages":""},"PeriodicalIF":10.7,"publicationDate":"2024-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11550092/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142633791","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}