Drug Resistance Updates最新文献

筛选
英文 中文
Engineering nanoplatforms of bacterial outer membrane vesicles to overcome cancer therapy resistance 细菌外膜囊泡的工程纳米平台克服癌症治疗耐药性
IF 15.8 1区 医学
Drug Resistance Updates Pub Date : 2025-07-18 DOI: 10.1016/j.drup.2025.101277
Qing-Qing Chai , Dan Li , Min Zhang , Yong-Wei Gu , Ai-Xue Li , Xin Wu , Xiao-Yan Liu , Ji-Yong Liu
{"title":"Engineering nanoplatforms of bacterial outer membrane vesicles to overcome cancer therapy resistance","authors":"Qing-Qing Chai ,&nbsp;Dan Li ,&nbsp;Min Zhang ,&nbsp;Yong-Wei Gu ,&nbsp;Ai-Xue Li ,&nbsp;Xin Wu ,&nbsp;Xiao-Yan Liu ,&nbsp;Ji-Yong Liu","doi":"10.1016/j.drup.2025.101277","DOIUrl":"10.1016/j.drup.2025.101277","url":null,"abstract":"<div><div>Resistance to cancer therapy is driven by physical barriers, tumor heterogeneity, selective therapeutic pressure, immunosuppressive tumor microenvironment (TME) and others. Bacterial outer membrane vesicles (OMVs) represent a promising nanotherapeutic platform to combat cancer therapy resistance. This review discusses the dual roles of OMVs in tumorigenesis and cancer therapy, highlighting their potential applications to enhance treatment efficacy. OMVs from pathogenic bacteria, such as <em>Fusobacterium nucleatum</em> and <em>Helicobacter pylori</em>, exacerbate chemoresistance by reshaping TME through hypoxia-induced metabolic reprogramming and immune evasion, while OMVs from some bacteria, such as probiotics, counteract immunosuppression by promoting cytotoxic T-cell infiltration and macrophage polarization. As bio-derived and conveniently engineered drug delivery platforms, OMVs maximize the synergetic anticancer effect by pathogen associated molecular patterns and the payloads. These functional payloads include siRNAs, cytotoxicity and molecular agents, and immune checkpoint inhibitors. Bacterial OMVs demonstrate unique advantages through their capacity to penetrate physical barriers, achieve tumor-specific targeting, activate immune responses, to overcome cancer therapy resistance. A successful example is the OMV-based nanoplatform with engineered OMVs co-delivering CD47-siRNA and doxorubicin to overcome drug resistance by inducing immunogenic cell death and dendritic cell activation of glioblastoma. Furthermore, OMV-based cancer vaccines presented with tumor antigens or hybridized with tumor-derived membranes enhance dendritic cell maturation and antigen-specific T-cell responses, reversing treatment resistance. By addressing challenges in mass production and safety concerns, OMVs-based platforms can be developed as powerful tools for more effective and personalized cancer treatments.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"83 ","pages":"Article 101277"},"PeriodicalIF":15.8,"publicationDate":"2025-07-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144665099","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
EMT and cancer stem cells: Drivers of therapy resistance and promising therapeutic targets EMT和癌症干细胞:治疗耐药的驱动因素和有希望的治疗靶点
IF 21.7 1区 医学
Drug Resistance Updates Pub Date : 2025-07-17 DOI: 10.1016/j.drup.2025.101276
Mingyang Jiang , Jinlong Wang , Yize Li , Ke Zhang , Tao Wang , Zhandong Bo , Shenyi Lu , Raquel Alarcón Rodríguez , Ruqiong Wei , Mingtao Zhu , Christophe Nicot , Gautam Sethi
{"title":"EMT and cancer stem cells: Drivers of therapy resistance and promising therapeutic targets","authors":"Mingyang Jiang ,&nbsp;Jinlong Wang ,&nbsp;Yize Li ,&nbsp;Ke Zhang ,&nbsp;Tao Wang ,&nbsp;Zhandong Bo ,&nbsp;Shenyi Lu ,&nbsp;Raquel Alarcón Rodríguez ,&nbsp;Ruqiong Wei ,&nbsp;Mingtao Zhu ,&nbsp;Christophe Nicot ,&nbsp;Gautam Sethi","doi":"10.1016/j.drup.2025.101276","DOIUrl":"10.1016/j.drup.2025.101276","url":null,"abstract":"<div><div>Cancer continues to be a primary cause of death, resulting in substantial mortality and illness globally. It remains a significant global health issue, greatly affecting morbidity and mortality across the world. Therapeutic resistance poses a major challenge to cancer treatments, acting as a significant barrier to the effectiveness of both standard and targeted therapies. This resistance develops through various mechanisms that allow tumor cells to adapt to and escape the damaging effects of chemotherapy, radiation, and targeted therapies. Ultimately, this leads to disease recurrence and progression. This review examines the dual roles of epithelial-mesenchymal transition (EMT) and cancer stem cells (CSCs) in promoting chemoresistance and metastasis. EMT is a dynamic and reversible biological process in which epithelial cells acquire mesenchymal characteristics, increasing their invasiveness and resistance to programmed cell death. CSCs are a subset of cancer cells with the ability to self-renew and play a crucial role in tumor relapse and resistance to treatment. EMT and CSCs are closely interconnected, collaboratively enhancing cancer cell plasticity, metastatic ability, and treatment resistance. The initiation of EMT in cancer cells can generate a CSC-like population, which promotes tumor recurrence and spread. This interaction highlights the importance of targeting both EMT and CSC pathways to develop more effective treatment strategies that address treatment resistance and prevent metastasis. Promising approaches include using natural substances, small molecules, and nanotechnology to block critical signaling pathways and interfere with resistance mechanisms. A more thorough understanding of the molecular factors underlying EMT and CSC plasticity is crucial for crafting personalized treatments that target tumor heterogeneity and improve clinical outcomes.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"83 ","pages":"Article 101276"},"PeriodicalIF":21.7,"publicationDate":"2025-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144725039","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Hypoxia-induced upregulation of HIF1A-AS3 promotes MSC transition to cancer-associated fibroblasts and confers drug resistance in gastric cancer 缺氧诱导的HIF1A-AS3上调可促进间充质干细胞向癌症相关成纤维细胞转化,并在胃癌中产生耐药性
IF 15.8 1区 医学
Drug Resistance Updates Pub Date : 2025-07-14 DOI: 10.1016/j.drup.2025.101275
Jiajin Xu , Shuo Fang , Xiaotong Dong , Chengdong Liang , Ruoyu Yang , Yang Zhao , Hailong Gu , Min Fu , Jiahui Zhang , Xiaoxin Zhang , Xu Zhang , Runbi Ji
{"title":"Hypoxia-induced upregulation of HIF1A-AS3 promotes MSC transition to cancer-associated fibroblasts and confers drug resistance in gastric cancer","authors":"Jiajin Xu ,&nbsp;Shuo Fang ,&nbsp;Xiaotong Dong ,&nbsp;Chengdong Liang ,&nbsp;Ruoyu Yang ,&nbsp;Yang Zhao ,&nbsp;Hailong Gu ,&nbsp;Min Fu ,&nbsp;Jiahui Zhang ,&nbsp;Xiaoxin Zhang ,&nbsp;Xu Zhang ,&nbsp;Runbi Ji","doi":"10.1016/j.drup.2025.101275","DOIUrl":"10.1016/j.drup.2025.101275","url":null,"abstract":"<div><div>Chemotherapy resistance is a major cause of poor prognosis in gastric cancer patients and tumor microenvironment plays a critical role in conferring chemotherapy resistance. As a dominant source of tumor stromal cells, mesenchymal stem cells (MSCs) exert pro-oncogenic activities when reprogrammed to a cancer-associated fibroblast (CAF) phenotype. The precise mechanisms for MSC reprogramming and their subsequent role in chemotherapy resistance have not been fully understood. Herein, we reported that HIF1A-AS3, a lncRNA that was highly expressed in tumor-promoting MSCs, was upregulated in tumor tissues and serum of gastric cancer patients and associated with poor prognosis. The upregulation of HIF1A-AS3 reprogramed MSCs to acquire the CAF phenotype, which consequently enhanced the resistance of gastric cancer cells to oxaliplatin. Mechanistically, hypoxia related transcription factor HIF-1α induced high expression of HIF1A-AS3 in MSCs. Then, HIF1A-AS3 competitively sponged miR-142–3p and miR-24–3p, leading to the upregulation of PROX1 (prospero-related homeobox protein 1) gene expression. This further promoted the nuclear translocation of β-catenin and the activation of β-catenin signaling pathway in MSCs, which critically regulated their transition to CAFs. Finally, targeted inhibition of HIF1A-AS3 in hypoxia-MSCs through exosome-mediated siRNA delivery significantly suppressed gastric cancer growth and improved chemosensitivity in mouse tumor models. Conclusively, hypoxia-induced HIF1A-AS3 upregulation reprograms MSCs to CAFs through the miR-142–3p/miR-24–3p/PROX1/β-catenin axis, thereby promoting chemotherapy resistance in gastric cancer, which uncovers a new molecular mechanism for MSCs transition to CAFs in gastric cancer and provides a new target for the diagnosis and targeted therapy of gastric cancer.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"82 ","pages":"Article 101275"},"PeriodicalIF":15.8,"publicationDate":"2025-07-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144665096","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
ZNF207-driven PRDX1 lactylation and NRF2 activation in regorafenib resistance and ferroptosis evasion znf207驱动的PRDX1乳酸化和NRF2激活在瑞非尼耐药和铁中毒逃避中的作用
IF 15.8 1区 医学
Drug Resistance Updates Pub Date : 2025-07-12 DOI: 10.1016/j.drup.2025.101274
Tianfeng Yang , Suyu Zhang , Kun Nie , Cheng Cheng , Xiuhong Peng , Jian Huo , Yanmin Zhang
{"title":"ZNF207-driven PRDX1 lactylation and NRF2 activation in regorafenib resistance and ferroptosis evasion","authors":"Tianfeng Yang ,&nbsp;Suyu Zhang ,&nbsp;Kun Nie ,&nbsp;Cheng Cheng ,&nbsp;Xiuhong Peng ,&nbsp;Jian Huo ,&nbsp;Yanmin Zhang","doi":"10.1016/j.drup.2025.101274","DOIUrl":"10.1016/j.drup.2025.101274","url":null,"abstract":"<div><div>Regorafenib (RGF) is a critical second-line therapy for advanced hepatocellular carcinoma (HCC) following disease progression on sorafenib; however, the rapid onset of RGF resistance poses a significant barrier to enhancing patient outcomes. In this study, CRISPR/Cas9 screening in RGF-treated HCC cells identified Zinc Finger Protein 207 (ZNF207) as a primary driver of resistance. Further analysis revealed that ZNF207 promotes resistance by inducing antioxidant responses that inhibit ferroptosis, a form of iron-dependent cell death. Mechanistically, ZNF207 facilitates the lactylation of peroxiredoxin 1 (PRDX1) at lysine 67, enhancing nuclear translocation and activation of nuclear factor erythroid 2–related factor 2 (NRF2), a master regulator of antioxidant pathways. This ZNF207-PRDX1-NRF2 pathway creates a ferroptosis-resistant, pro-survival environment under RGF treatment, enabling HCC cells to evade cell death. Functional assays demonstrated that ZNF207 knockdown significantly enhances RGF sensitivity by restoring ferroptosis, with additional findings showing that disrupting PRDX1 lactylation or NRF2 activity similarly reverses resistance. Together, these findings establish a critical link between protein lactylation and RGF resistance, positioning the ZNF207-PRDX1-NRF2 axis as a promising therapeutic target to enhance treatment efficacy in HCC. The implications of this research extend beyond HCC, indicating that targeting ferroptosis-suppressive pathways may offer a broader approach to overcoming resistance in various cancers.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"82 ","pages":"Article 101274"},"PeriodicalIF":15.8,"publicationDate":"2025-07-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144613303","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The multifaceted contributions of cancer-associated fibroblasts to drug resistance in primary and metastatic tumors 癌症相关成纤维细胞对原发性和转移性肿瘤耐药的多方面贡献
IF 15.8 1区 医学
Drug Resistance Updates Pub Date : 2025-07-01 DOI: 10.1016/j.drup.2025.101273
Xinhao Zhang , Yuhang Wang , Wenming Cui , Danyang Li , Junmin Song , Zhen Li , Ying Liu , Shuaixi Yang
{"title":"The multifaceted contributions of cancer-associated fibroblasts to drug resistance in primary and metastatic tumors","authors":"Xinhao Zhang ,&nbsp;Yuhang Wang ,&nbsp;Wenming Cui ,&nbsp;Danyang Li ,&nbsp;Junmin Song ,&nbsp;Zhen Li ,&nbsp;Ying Liu ,&nbsp;Shuaixi Yang","doi":"10.1016/j.drup.2025.101273","DOIUrl":"10.1016/j.drup.2025.101273","url":null,"abstract":"<div><div>Drug resistance remains a formidable barrier in modern oncology, undermining the efficacy of the current therapeutic regimens. As pivotal stromal constituents within tumor ecosystems, cancer-associated fibroblasts (CAFs) have emerged as critical mediators of treatment resistance through multifaceted mechanisms. In this review, we summarize the historical progression of research on CAFs and drug resistance, and highlight the recent discoveries related to CAF biomarkers and their functions associated with drug resistance. Furthermore, we discuss the relationship between CAF heterogeneity, secretion, autophagy, and senescence and their contributions to the evolution of drug resistance. Additionally, we provide a detailed explanation of how CAFs contribute to the development of drug resistance in primary tumors, including mechanisms such as immune suppression and evasion, promotion of tumor stemness, angiogenesis, extracellular matrix remodeling, and metabolic reprogramming. We also explored the role of CAFs in metastatic tumors and their association with drug resistance at various metastatic sites, including lymph nodes, brain, lungs, peritoneum, bone, and liver. Finally, we summarize the advancements in clinical trials targeting CAFs, the emerging research on potential therapeutic targets, and anticipating future trends in this area.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"82 ","pages":"Article 101273"},"PeriodicalIF":15.8,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144562800","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Metabolic reprogramming of tumor-associated macrophages via adenosine-A2AR signaling drives cross-resistance in non-small cell lung cancer 肿瘤相关巨噬细胞通过腺苷- a2ar信号的代谢重编程驱动非小细胞肺癌的交叉耐药
IF 15.8 1区 医学
Drug Resistance Updates Pub Date : 2025-06-30 DOI: 10.1016/j.drup.2025.101272
Wen Ding , Jianshan Mo , Yingxue Su , Qiyi Zhang , Danyuan Sun , Xiangchao Yao , Guopin Liu , Jiangling Ye , Yanle Wu , Menghan Xue , Peibin Yue , Jinjian Lu , Jian Zhang , Yanyu Shi , Wenhao Hu , Kai Zhu , Yandong Wang , Xiaolei Zhang
{"title":"Metabolic reprogramming of tumor-associated macrophages via adenosine-A2AR signaling drives cross-resistance in non-small cell lung cancer","authors":"Wen Ding ,&nbsp;Jianshan Mo ,&nbsp;Yingxue Su ,&nbsp;Qiyi Zhang ,&nbsp;Danyuan Sun ,&nbsp;Xiangchao Yao ,&nbsp;Guopin Liu ,&nbsp;Jiangling Ye ,&nbsp;Yanle Wu ,&nbsp;Menghan Xue ,&nbsp;Peibin Yue ,&nbsp;Jinjian Lu ,&nbsp;Jian Zhang ,&nbsp;Yanyu Shi ,&nbsp;Wenhao Hu ,&nbsp;Kai Zhu ,&nbsp;Yandong Wang ,&nbsp;Xiaolei Zhang","doi":"10.1016/j.drup.2025.101272","DOIUrl":"10.1016/j.drup.2025.101272","url":null,"abstract":"<div><div>Immunosuppression within the tumor microenvironment (TME) is frequently associated with chemoresistance. However, the mechanisms by which chemoresistance promotes immune evasion and impairs subsequent immunotherapy remain poorly understood, underscoring the urgent need for novel therapeutic strategies to counteract these effects. In this study, we observed that tumors exhibit cross-resistance to immunotherapy following chemoresistance in a non-small cell lung cancer (NSCLC) mouse model. The aberrant accumulation of tumor-associated macrophages (TAMs) and extracellular adenosine (Ado) were identified as mediators of immunosuppression, fostering cross-resistance to immunotherapy in the context of chemoresistance. Mechanistically, selective activation of the Ado/A<sub>2A</sub>R signaling pathway induced metabolic reprogramming of TAMs, thereby creating an immunosuppressive niche in cross-resistant NSCLC. Based on these findings, we designed a novel selective A<sub>2A</sub>R inhibitor DL082 and explored its therapeutic potential for treating cross-resistant NSCLC. The combination of DL082 with an anti-PD-L1 antibody significantly enhanced immune activation and inhibited tumor progression in cross-resistant NSCLC. These findings elucidate the specific mechanisms underlying cross-resistance between chemotherapy and immunotherapy in NSCLC and propose targeting the Ado-TAM axis as a potential strategy for overcoming resistance in NSCLC therapy.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"82 ","pages":"Article 101272"},"PeriodicalIF":15.8,"publicationDate":"2025-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144563199","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Corrigendum to “NDRG1 overcomes resistance to immunotherapy of pancreatic ductal adenocarcinoma through inhibiting ATG9A-dependent degradation of MHC-I” [Drug Resist. Updates 73 (2024) 101040] “NDRG1通过抑制atg9a依赖性MHC-I降解来克服对胰腺导管腺癌免疫治疗的耐药性”[耐药]的更正。更新73 (2024)101040]
IF 15.8 1区 医学
Drug Resistance Updates Pub Date : 2025-06-13 DOI: 10.1016/j.drup.2025.101269
Zhiheng Zhang , Bojiao Song , Haowei Wei , Yang Liu , Wenjie Zhang , Yuhong Yang , Beicheng Sun
{"title":"Corrigendum to “NDRG1 overcomes resistance to immunotherapy of pancreatic ductal adenocarcinoma through inhibiting ATG9A-dependent degradation of MHC-I” [Drug Resist. Updates 73 (2024) 101040]","authors":"Zhiheng Zhang ,&nbsp;Bojiao Song ,&nbsp;Haowei Wei ,&nbsp;Yang Liu ,&nbsp;Wenjie Zhang ,&nbsp;Yuhong Yang ,&nbsp;Beicheng Sun","doi":"10.1016/j.drup.2025.101269","DOIUrl":"10.1016/j.drup.2025.101269","url":null,"abstract":"","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"82 ","pages":"Article 101269"},"PeriodicalIF":15.8,"publicationDate":"2025-06-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144289794","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Corrigendum to “A novel platinum(IV) prodrug, gramine-Pt(IV) enhances chemoimmunotherapy by activating cGAS-STING and modulating TGF-β-MHC-I axis” [Drug Resist. Updates 81 (2025) 101252] “一种新型铂(IV)前药,gramine-Pt(IV)通过激活cGAS-STING和调节TGF-β-MHC-I轴来增强化学免疫治疗”的勘误表。更新81 (2025)101252]
IF 15.8 1区 医学
Drug Resistance Updates Pub Date : 2025-06-13 DOI: 10.1016/j.drup.2025.101268
Bowen Ding , Xiaomeng Liu , Zhe Li , Xinru Xie , Jiaqi Li , Jiaqian Wang , Shouyi Li , Pengyu Wang , Yongjie Xie , Xiaoqing Ma , Hongwei Wang , Chengzhi Xie , Xin Qiao , Yumin Wang , Jingyuan Xu , Yukuan Feng , Jihui Hao
{"title":"Corrigendum to “A novel platinum(IV) prodrug, gramine-Pt(IV) enhances chemoimmunotherapy by activating cGAS-STING and modulating TGF-β-MHC-I axis” [Drug Resist. Updates 81 (2025) 101252]","authors":"Bowen Ding ,&nbsp;Xiaomeng Liu ,&nbsp;Zhe Li ,&nbsp;Xinru Xie ,&nbsp;Jiaqi Li ,&nbsp;Jiaqian Wang ,&nbsp;Shouyi Li ,&nbsp;Pengyu Wang ,&nbsp;Yongjie Xie ,&nbsp;Xiaoqing Ma ,&nbsp;Hongwei Wang ,&nbsp;Chengzhi Xie ,&nbsp;Xin Qiao ,&nbsp;Yumin Wang ,&nbsp;Jingyuan Xu ,&nbsp;Yukuan Feng ,&nbsp;Jihui Hao","doi":"10.1016/j.drup.2025.101268","DOIUrl":"10.1016/j.drup.2025.101268","url":null,"abstract":"","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"82 ","pages":"Article 101268"},"PeriodicalIF":15.8,"publicationDate":"2025-06-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144289795","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Molecular mechanisms of acquired resistance to EGFR tyrosine kinase inhibitors in non-small cell lung cancer 非小细胞肺癌对EGFR酪氨酸激酶抑制剂获得性耐药的分子机制
IF 15.8 1区 医学
Drug Resistance Updates Pub Date : 2025-06-09 DOI: 10.1016/j.drup.2025.101266
Zhenfang Du , Hongfei Kan , Jinghan Sun , Yue Liu , Jiahui Gu , Shugela Akemujiang , Yudi Zou , Lufan Jiang , Qinzhuo Wang , Chen Li , Lei Luo , Yunkai Zhang , Hong Fan , Pengfei Luo , Bo Wang
{"title":"Molecular mechanisms of acquired resistance to EGFR tyrosine kinase inhibitors in non-small cell lung cancer","authors":"Zhenfang Du ,&nbsp;Hongfei Kan ,&nbsp;Jinghan Sun ,&nbsp;Yue Liu ,&nbsp;Jiahui Gu ,&nbsp;Shugela Akemujiang ,&nbsp;Yudi Zou ,&nbsp;Lufan Jiang ,&nbsp;Qinzhuo Wang ,&nbsp;Chen Li ,&nbsp;Lei Luo ,&nbsp;Yunkai Zhang ,&nbsp;Hong Fan ,&nbsp;Pengfei Luo ,&nbsp;Bo Wang","doi":"10.1016/j.drup.2025.101266","DOIUrl":"10.1016/j.drup.2025.101266","url":null,"abstract":"<div><div>Tyrosine kinase inhibitors (TKIs) have revolutionized the management of epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC). Despite the initially favorable outcomes, these patients inevitably acquire resistance to EGFR-TKIs. The molecular mechanism of the acquired resistance is highly complex and heterogeneous, and can be generalized as three categories, including EGFR pathway reactivation (re-engagement of EGFR downstream), EGFR pathway bypass (adoption of a parallel pathway to re-engage the downstream transcriptional oncogenic output of the original EGFR), and EGFR pathway indifference (acquirement of a cellular state alternate to the original EGFR-driven output). This review summarizes the recent progress on the identification and understanding of the acquired resistance mechanisms to EGFR-TKIs in patients with NSCLC. The potential strategies to delay or overcome the acquired resistance are also discussed.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"82 ","pages":"Article 101266"},"PeriodicalIF":15.8,"publicationDate":"2025-06-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144261985","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Exosome-derived lnc-FAM72D-3 promotes lenvatinib resistance by remodeling hepatocellular carcinoma cytoskeleton via MBNL1/FAK axis 外泌体衍生的lnc-FAM72D-3通过MBNL1/FAK轴重塑肝癌细胞骨架,促进lenvatinib耐药
IF 15.8 1区 医学
Drug Resistance Updates Pub Date : 2025-06-07 DOI: 10.1016/j.drup.2025.101271
Mingbo Cao , Yuxuan Li , Xiaorui Su , Yongchang Tang , Yupeng Ren , Jing Luo , Feng Yuan , Gaoyuan Yang , Zhiwei He , Zheng Shi , Ziyi Hu , Guirong Liang , Qi Zhang , Meihai Deng , Zhicheng Yao , Nan Lin
{"title":"Exosome-derived lnc-FAM72D-3 promotes lenvatinib resistance by remodeling hepatocellular carcinoma cytoskeleton via MBNL1/FAK axis","authors":"Mingbo Cao ,&nbsp;Yuxuan Li ,&nbsp;Xiaorui Su ,&nbsp;Yongchang Tang ,&nbsp;Yupeng Ren ,&nbsp;Jing Luo ,&nbsp;Feng Yuan ,&nbsp;Gaoyuan Yang ,&nbsp;Zhiwei He ,&nbsp;Zheng Shi ,&nbsp;Ziyi Hu ,&nbsp;Guirong Liang ,&nbsp;Qi Zhang ,&nbsp;Meihai Deng ,&nbsp;Zhicheng Yao ,&nbsp;Nan Lin","doi":"10.1016/j.drup.2025.101271","DOIUrl":"10.1016/j.drup.2025.101271","url":null,"abstract":"<div><div>Lenvatinib resistance (LR) profoundly exacerbates the prognosis of patients afflicted with advanced hepatocellular carcinoma (HCC). As pivotal mediators of intercellular communication, exosomes have been implicated in the development of LR. Nonetheless, the precise contributions of exosome-derived long non-coding RNAs (lncRNAs) to this phenomenon remain inadequately elucidated. Our prior investigations identified that lnc-FAM72D-3 is markedly up-regulated in the serum exosomes of HCC patients, yet its specific functions and underlying mechanisms remain only partially defined. In this study, we established lenvatinib-resistant HCC cell lines and organoids and demonstrated, through rigorous in vitro and in vivo experiments, that exosome-derived lnc-FAM72D-3 facilitates HCC progression and contributes to the phenomenon of LR. Mechanistically, lnc-FAM72D-3 augments the affinity of the E3 ubiquitin ligase HECTD3 for MBNL1, inciting lysine 48-linked ubiquitination and subsequent degradation of MBNL1. This degradation diminishes the interaction between MBNL1 and focal adhesion kinase (FAK), precipitating the de-nucleation of FAK and its activation by phosphorylation. The activated FAK subsequently reorganizes the cytoskeleton, markedly enhancing the proliferation, invasion, and stemness of HCC cells, thereby fostering LR. In summary, this investigation offers novel mechanistic insights into the regulatory role of exosomal lncRNAs in LR and posits a potential therapeutic strategy aimed at mitigating LR in patients with HCC.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"82 ","pages":"Article 101271"},"PeriodicalIF":15.8,"publicationDate":"2025-06-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144280572","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
相关产品
×
本文献相关产品
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信