癌症耐药(英文)最新文献

筛选
英文 中文
Distinct adaptive strategies to cisplatin, vinblastine and gemcitabine in a panel of chemoresistant bladder cancer cell lines. 一组化疗耐药膀胱癌细胞系对顺铂、长春碱和吉西他滨的不同适应策略
IF 4.6
癌症耐药(英文) Pub Date : 2025-09-12 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.95
Monika Cuprych-Belter, Agnieszka Łupicka-Słowik, Artur Anisiewicz, Martin Michaelis, Jindrich Cinatl, Mateusz Psurski
{"title":"Distinct adaptive strategies to cisplatin, vinblastine and gemcitabine in a panel of chemoresistant bladder cancer cell lines.","authors":"Monika Cuprych-Belter, Agnieszka Łupicka-Słowik, Artur Anisiewicz, Martin Michaelis, Jindrich Cinatl, Mateusz Psurski","doi":"10.20517/cdr.2025.95","DOIUrl":"10.20517/cdr.2025.95","url":null,"abstract":"<p><p><b>Aim:</b> Urinary bladder cancer (UBC) often develops chemoresistance, reducing treatment effectiveness. This study aimed to investigate diverse molecular mechanisms underlying acquired resistance by establishing and characterizing a comprehensive panel of UBC cell lines resistant to common chemotherapeutics. <b>Methods:</b> Fifteen UBC cell lines were examined: three parental lines (RT-112, TCC-SUP, UMUC-3) and twelve derived sublines adapted to cisplatin, vinblastine, or gemcitabine. Drug sensitivity was assessed using the SRB assay. Resistance mechanisms were explored via quantitative real-time PCR (targeting genes including <i>ABCB1</i>, <i>dCK</i>, <i>hENT1</i>, <i>ECHDC1</i>, <i>TUBB3</i>), Western blotting (assessing proteins such as p21, Cyclin B, and Mcl-1), and biochemical assessment of glutathione levels and redox state. <b>Results:</b> The adapted sublines exhibited distinct resistance profiles and cross-resistance patterns. Gene expression and protein analyses revealed drug- and lineage-specific alterations, involving factors such as p21, Cyclin B, and Mcl-1. Changes in glutathione metabolism were also associated with resistance. Notably, no single, universal mechanism accounted for resistance across the entire panel. <b>Conclusion:</b> UBC cells develop diverse, context-dependent adaptive strategies to resist cisplatin, vinblastine, and gemcitabine. These findings highlight the complexity of chemoresistance mechanisms. The characterized cell line panel represents a valuable resource for future studies aimed at understanding and overcoming drug resistance in bladder cancer, suggesting that personalized therapeutic approaches may be necessary.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"49"},"PeriodicalIF":4.6,"publicationDate":"2025-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12462402/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145187647","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}
引用次数: 0
Immune checkpoint inhibitors in hepatocellular carcinoma therapy: resistance mechanisms, liver transplantation challenges and management strategies. 免疫检查点抑制剂在肝细胞癌治疗中的应用:耐药机制、肝移植挑战和管理策略。
IF 4.6
癌症耐药(英文) Pub Date : 2025-09-11 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.120
Yutao Chen, Desheng Chen, Zhixing Liang, Haoyuan Yu, Haobin Sun, Yongwei Hu, Peng Jiang, Mingshen Zhang, Linsen Ye, Hua Li
{"title":"Immune checkpoint inhibitors in hepatocellular carcinoma therapy: resistance mechanisms, liver transplantation challenges and management strategies.","authors":"Yutao Chen, Desheng Chen, Zhixing Liang, Haoyuan Yu, Haobin Sun, Yongwei Hu, Peng Jiang, Mingshen Zhang, Linsen Ye, Hua Li","doi":"10.20517/cdr.2025.120","DOIUrl":"10.20517/cdr.2025.120","url":null,"abstract":"<p><p>Hepatocellular carcinoma (HCC) poses a significant clinical burden due to its aggressive nature, profound tumor heterogeneity, and limited therapeutic efficacy. While immune checkpoint inhibitors (ICIs) have revolutionized treatment paradigms and demonstrated considerable promise, the emergence of resistance mechanisms has posed a critical challenge in contemporary clinical oncology. The accelerated development of novel agents and innovative combination strategies has further complicated this resistance landscape. In this review, we present a unique and comprehensive analysis of ICI resistance mechanisms in HCC by integrating insights into primary resistance, acquired resistance, and host-related factors. Building upon this mechanistic framework, we explore emerging therapeutic strategies to overcome ICI resistance. Furthermore, we evaluate the dual role of ICIs in HCC management - serving as a neoadjuvant therapy for transplant candidates while simultaneously posing risks of post-transplant rejection. By bridging preclinical discoveries with clinical realities, this analysis aims to inform rational therapeutic design and optimize immuno-oncology trials for HCC patients.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"48"},"PeriodicalIF":4.6,"publicationDate":"2025-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12462397/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145187704","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}
引用次数: 0
Integrating genomic mutations and tumor-infiltrating lymphocytes improves prediction of response to trastuzumab-based adjuvant therapy in patients with HER2-positive breast cancer. 整合基因组突变和肿瘤浸润淋巴细胞可提高her2阳性乳腺癌患者对曲妥珠单抗辅助治疗反应的预测。
IF 4.6
癌症耐药(英文) Pub Date : 2025-09-08 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.133
Shuangshuang Lu, Yuliang Zhang, Yiwei Tong, Lan Shu, Renhong Huang, Yijin Gu, Chaofu Wang, Jianfeng Li, Kunwei Shen, Lei Dong, Xiaosong Chen
{"title":"Integrating genomic mutations and tumor-infiltrating lymphocytes improves prediction of response to trastuzumab-based adjuvant therapy in patients with HER2-positive breast cancer.","authors":"Shuangshuang Lu, Yuliang Zhang, Yiwei Tong, Lan Shu, Renhong Huang, Yijin Gu, Chaofu Wang, Jianfeng Li, Kunwei Shen, Lei Dong, Xiaosong Chen","doi":"10.20517/cdr.2025.133","DOIUrl":"10.20517/cdr.2025.133","url":null,"abstract":"<p><p><b>Aim:</b> Resistance to trastuzumab remains a major barrier to cure in early-stage HER2-positive breast cancer (HER2+ BC). We investigated the impact of genomic alterations and tumor-infiltrating lymphocyte (TIL) density on treatment resistance and survival outcomes. <b>Methods:</b> We retrospectively analyzed 315 patients with HER2+ BC who received adjuvant trastuzumab at Ruijin Hospital (2009-2019). Whole-exome sequencing and TIL scoring were performed on surgical specimens, and clinical and pathological data were collected. The Cancer Genome Atlas (TCGA) cohort was used for external validation. Genomic alterations and TIL density were compared between trastuzumab-sensitive and -resistant tumors. Survival analyses were conducted to identify prognostic biomarkers. <b>Results:</b> After a median follow-up of 109.3 months, 67 tumors (21.3%) were trastuzumab-resistant, exhibiting lower TIL density (mean 19.8% <i>vs.</i> 26.3%, <i>P</i> = 0.001), higher mutation frequencies in <i>FLG</i>, <i>MAP1A</i>, <i>BRCA1</i>, <i>PTPRD</i>, <i>PAPPA2</i>, <i>NCOR2</i>, <i>FBXW7</i>, <i>MYH7</i>, and <i>VCAN</i>, and more frequent alterations in the TP53/NOTCH pathways compared with sensitive tumors (all <i>P</i> < 0.05). A 15-gene trastuzumab response-associated gene (TRAG) signature independently predicted poorer disease-free survival (DFS) in both our cohort (HR, 3.57, <i>P</i> < 0.001) and the TCGA cohort (HR, 4.99, <i>P</i> = 0.037). A high copy number alteration burden was associated with worse overall survival (HR, 2.49, <i>P</i> = 0.043), whereas TIL density > 10% was associated with improved DFS (HR, 2.44, <i>P</i> = 0.003). A prognostic model integrating tumor size, nodal status, estrogen receptor status, TILs, and the TRAG signature showed strong discriminatory power (c-index 0.743 in the training set; 0.915 in the validation set). <b>Conclusion:</b> Genomic alterations and reduced TIL density underpin trastuzumab resistance. The novel TRAG signature and integrated prognostic model enhance risk stratification and may guide personalized adjuvant therapy in early-stage HER2+ BC.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"47"},"PeriodicalIF":4.6,"publicationDate":"2025-09-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12462396/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145187688","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}
引用次数: 0
Targeting the hypoxia signaling pathway with nanomedicine to reverse immunotherapy resistance. 纳米药物靶向缺氧信号通路逆转免疫治疗耐药。
IF 4.6
癌症耐药(英文) Pub Date : 2025-09-02 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.132
Xiaoliang Cheng, Peixing Wang, Hongqiang Lyu, Yonghyun Lee, Juyoung Yoon, Haiyan Dong
{"title":"Targeting the hypoxia signaling pathway with nanomedicine to reverse immunotherapy resistance.","authors":"Xiaoliang Cheng, Peixing Wang, Hongqiang Lyu, Yonghyun Lee, Juyoung Yoon, Haiyan Dong","doi":"10.20517/cdr.2025.132","DOIUrl":"10.20517/cdr.2025.132","url":null,"abstract":"<p><p>Immunotherapy has emerged as a major therapeutic strategy for cancer; however, immunotherapy resistance remains a significant challenge. Hypoxia, a key hallmark of the tumor microenvironment resulting from the imbalance between the high oxygen demand of rapidly proliferating cancer cells and the limited supply from abnormal blood vessels, plays a central role in driving immunotherapy resistance. Hypoxia-inducible factor-1α (HIF-1α) and its downstream signaling pathways contribute to this resistance by promoting macrophage polarization toward the protumorigenic M2 phenotype, inducing T cell exhaustion, facilitating immune evasion, enhancing angiogenesis, and activating other resistance mechanisms. The review highlights the mechanisms by which hypoxia regulates resistance to immunotherapy and provides a comprehensive overview of nanotechnology-based strategies designed to counteract hypoxia-induced resistance. Finally, the prospects and challenges of translating nanomedicine-based drug delivery systems into clinical practice for overcoming immunotherapy resistance are outlined.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"46"},"PeriodicalIF":4.6,"publicationDate":"2025-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12462398/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145187714","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}
引用次数: 0
Cryptotanshinone differentially induces cell death in ATP6V0D1-deficient pancreatic cancer cells. 隐丹参酮诱导atp6v0d1缺陷胰腺癌细胞死亡的差异
IF 4.6
癌症耐药(英文) Pub Date : 2025-08-27 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.103
Fangquan Chen, Junhao Lin, Xiutao Cai, Hu Tang, Shengfeng Li, Ruirui Liang, Rui Kang, Zhenhui Zhang, Daolin Tang, Jiao Liu
{"title":"Cryptotanshinone differentially induces cell death in <i>ATP6V0D1</i>-deficient pancreatic cancer cells.","authors":"Fangquan Chen, Junhao Lin, Xiutao Cai, Hu Tang, Shengfeng Li, Ruirui Liang, Rui Kang, Zhenhui Zhang, Daolin Tang, Jiao Liu","doi":"10.20517/cdr.2025.103","DOIUrl":"10.20517/cdr.2025.103","url":null,"abstract":"<p><p><b>Aim:</b> Dysregulation of tumor-suppressive pathways can lead to constitutive activation of multiple oncogenic signaling cascades. Such overactivation makes cancer cells highly dependent on these pathways, creating potential therapeutic vulnerabilities. Based on our previous findings and current data, genetic knockout of ATPase H<sup>+</sup> transporting V0 subunit D1 (ATP6V0D1) - a key mediator of alkaliptosis - induces hyperactivation of oncogenic pathways, including signal transducer and activator of transcription 3 (STAT3)-mediated lysosomal pH regulation and AKT serine/threonine kinase (AKT) signaling. It also alters cellular responses to cryptotanshinone therapy. This study aimed to investigate how <i>ATP6V0D1</i> deficiency reshapes oncogenic signaling networks and cellular heterogeneity in pancreatic ductal adenocarcinoma (PDAC), while evaluating therapeutic strategies that exploit alkaliptosis-related vulnerabilities. <b>Methods:</b> <i>ATP6V0D1</i>-deficient SW1990 and MIAPaCa2 cells were generated via gene knockdown. Cell viability and death following various treatments were assessed using CCK-8 and propidium iodide assays. Transcriptomic analysis was conducted to identify feedback signaling pathways, while Western blotting was used to measure expression of signaling proteins. Macropinocytosis was evaluated by TRITC-dextran uptake. Additionally, The Cancer Dependency Map (DepMap) database was analyzed to explore background differences between SW1990 and MIAPaCa2 cells. <b>Results:</b> <i>ATP6V0D1</i> deletion led to overactivation of STAT3-mediated lysosomal pH regulation and AKT signaling; inhibition of these pathways restored alkaliptosis. Notably, cryptotanshinone selectively induced cell death in <i>ATP6V0D1</i>-deficient MIAPaCa2 cells but not SW1990 cells. Resistance in SW1990 cells was mediated by FGFR2 upregulation, which was reversed upon FGFR2 inhibition. <b>Conclusion:</b> <i>ATP6V0D1</i> deficiency drives PDAC progression via dual mechanisms: compensatory oncogenic signaling (STAT3/AKT) and FGFR2-mediated cellular heterogeneity. While targeting these pathways may offer therapeutic potential, tumor heterogeneity remains a major clinical challenge.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"45"},"PeriodicalIF":4.6,"publicationDate":"2025-08-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12462395/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145187630","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}
引用次数: 0
Resistance signatures manifested in early drug response across cancer types and species. 耐药特征表现在癌症类型和物种的早期药物反应中。
IF 4.6
癌症耐药(英文) Pub Date : 2025-08-26 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.112
Cole Ruoff, Allison Mitchell, Priya Mondal, Vishaka Gopalan, Arashdeep Singh, Michael Gottesman, Sridhar Hannenhalli
{"title":"Resistance signatures manifested in early drug response across cancer types and species.","authors":"Cole Ruoff, Allison Mitchell, Priya Mondal, Vishaka Gopalan, Arashdeep Singh, Michael Gottesman, Sridhar Hannenhalli","doi":"10.20517/cdr.2025.112","DOIUrl":"10.20517/cdr.2025.112","url":null,"abstract":"<p><p><b>Aim:</b> Growing evidence points to non-genetic mechanisms underlying long-term resistance to cancer therapies. These mechanisms involve pre-existing or therapy-induced transcriptional cell states that confer resistance. However, the relationship between early transcriptional responses to treatment and the eventual emergence of resistant states remains poorly understood. Furthermore, it is unclear whether such early resistance-associated transcriptional responses are evolutionarily conserved. In this study, we examine the similarity between early transcriptional responses and long-term resistant states, assess their clinical relevance, and explore their evolutionary conservation across species. <b>Methods:</b> We integrated datasets on early drug responses and long-term resistance from multiple cancer cell lines, bacteria, and yeast to identify early transcriptional changes predictive of long-term resistance and assess their evolutionary conservation. Using genome-wide CRISPR-Cas9 knockout screens, we evaluated the impact of genes associated with resistant transcriptional states on drug sensitivity. Clinical datasets were analyzed to explore the prognostic value of the identified resistance-associated gene signatures. <b>Results:</b> We found that transcriptional states observed in drug-naive cells and shortly after treatment overlapped with those seen in fully resistant populations. Some of these shared features appear to be evolutionarily conserved. Knockout of genes marking resistant states sensitized ovarian cancer cells to Prexasertib. Moreover, early resistance gene signatures effectively distinguished therapy responders from non-responders in multiple clinical cancer trials and differentiated premalignant breast lesions that progressed to malignancy from those that remained benign. <b>Conclusion:</b> Early cellular transcriptional responses to therapy exhibit key similarities to fully resistant states across different drugs, cancer types, and species. Gene signatures defining these early resistance states have prognostic value in clinical settings.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"44"},"PeriodicalIF":4.6,"publicationDate":"2025-08-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12462399/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145187645","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}
引用次数: 0
New advances in understanding the mechanisms and treatment challenges of ALK-targeted therapy resistance in lung cancer. 肺癌alk靶向治疗耐药机制和治疗挑战的新进展。
IF 4.6
癌症耐药(英文) Pub Date : 2025-08-25 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.122
Mengle Long, Shixuan Peng, Qingyang Wen, Zhijian Yin, Xinwen Zhang, Haoyu Tan, Yun Xu, Yongjun Wu
{"title":"New advances in understanding the mechanisms and treatment challenges of ALK-targeted therapy resistance in lung cancer.","authors":"Mengle Long, Shixuan Peng, Qingyang Wen, Zhijian Yin, Xinwen Zhang, Haoyu Tan, Yun Xu, Yongjun Wu","doi":"10.20517/cdr.2025.122","DOIUrl":"10.20517/cdr.2025.122","url":null,"abstract":"<p><p>Despite the development of various effective anaplastic lymphoma kinase tyrosine kinase inhibitors (ALK-TKIs), therapeutic resistance remains a major challenge. Both on-target and off-target mechanisms have been identified as key contributors to resistance. With the popularization of genetic testing and the development of precision therapies, the prognosis and survival of patients with ALK-positive non-small cell lung cancer (NSCLC) have improved. However, even with second- and third-generation ALK-TKIs, overcoming resistance remains difficult. Resistance frequently arises during approved treatments, underscoring the need for further research to elucidate the molecular events and resistance mechanisms associated with ALK-positive lung cancer. The discovery of anaplastic lymphoma kinase (ALK) rearrangement as an actionable oncogenic driver in NSCLC has established a biomarker-driven treatment paradigm for advanced disease. This article summarizes current knowledge of the mechanisms of resistance to ALK-targeted therapy in lung cancer, including both primary and acquired mechanisms, treatment strategies following resistance, recent therapeutic advances, and the impact of the immune system and tumor microenvironment. A deeper understanding of ALK-targeted therapy resistance is critical for developing new treatment strategies and may provide important insights to guide the diagnosis, treatment, and management of patients with resistant ALK<sup>+</sup> lung cancer.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"43"},"PeriodicalIF":4.6,"publicationDate":"2025-08-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12378566/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144980620","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}
引用次数: 0
ROS1 mutations promote an immunosuppressive tumor microenvironment via MYC to confer immune evasion in head and neck cancer. ROS1突变通过MYC促进免疫抑制肿瘤微环境,从而导致头颈癌的免疫逃避。
IF 4.6
癌症耐药(英文) Pub Date : 2025-08-22 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.124
Chao Fang, Qin Zhang, Rui Fang, Ying Li, Jing Bai, Xiaojing Huang, Jingting Lu, Dongsheng Chen, Yanxiang Zhang, Zuhong Chen
{"title":"<i>ROS1</i> mutations promote an immunosuppressive tumor microenvironment via <i>MYC</i> to confer immune evasion in head and neck cancer.","authors":"Chao Fang, Qin Zhang, Rui Fang, Ying Li, Jing Bai, Xiaojing Huang, Jingting Lu, Dongsheng Chen, Yanxiang Zhang, Zuhong Chen","doi":"10.20517/cdr.2025.124","DOIUrl":"10.20517/cdr.2025.124","url":null,"abstract":"<p><p><b>Aim:</b> Immune checkpoint inhibitors (ICIs) have transformed cancer therapy; however, their efficacy in head and neck cancer (HNC) remains limited, with only a minority of patients achieving durable responses. Understanding the molecular mechanisms underlying ICI resistance in HNC is therefore crucial. <b>Methods:</b> We conducted an integrative analysis of genomic, transcriptomic, and clinical data from 139 ICI-treated HNC patients (MSKCC cohort) and 502 treatment-naïve HNC cases (TCGA cohort). <i>ROS1</i> mutation status, tumor mutational burden (TMB), neoantigen load, immune cell infiltration (via CIBERSORT), and immune-related gene expression were evaluated. Gene set enrichment analysis (GSEA) was performed to identify dysregulated pathways. Survival outcomes were assessed using Kaplan-Meier analysis and Cox regression, with statistical significance defined as <i>P</i> < 0.05. <b>Results:</b> Patients harboring <i>ROS1</i> mutations exhibited significantly poorer outcomes following ICI therapy, with shorter median overall survival [OS: 5.0 <i>vs.</i> 11.0 months, hazard ratio (HR) = 3.22, 95%CI: 1.26-8.19, <i>P</i> = 0.011] compared to <i>ROS1</i> wild-type counterparts. Multivariate analysis confirmed <i>ROS1</i> mutation as an independent predictor of poor OS in ICI-treated patients (HR = 4.78, 95%CI: 1.70-13.43, <i>P</i> = 0.003). In contrast, <i>ROS1</i> mutations showed no prognostic significance in the treatment-naïve TCGA-HNC cohort (<i>P</i> = 0.26), confirming their role as a predictive (not prognostic) biomarker for ICI response. Interestingly, despite exhibiting higher TMB and neoantigen levels, <i>ROS1</i>-mutant patients showed inferior survival, underscoring the context-dependent limitations of TMB as a predictive biomarker. Mechanistically, <i>ROS1</i>-mutant tumors displayed an immunosuppressive tumor microenvironment characterized by diminished CD8<sup>+</sup> T cell infiltration, attenuated interferon-γ signaling, and downregulation of immune-related genes (<i>CXCL9</i>, <i>CXCL10</i>, <i>IFNG</i>, <i>PD-L1</i>). GSEA revealed enrichment of <i>MYC</i> pathway activity in <i>ROS1</i>-mutant tumors, which suppressed antigen presentation and T cell activation pathways. <b>Conclusion:</b> <i>ROS1</i> mutations drive ICI resistance in HNC by promoting an immunosuppressive TME via <i>MYC</i>-mediated transcriptional reprogramming, impairing antigen presentation and T cell function. Incorporating <i>ROS1</i> status into biomarker panels may improve patient stratification and guide combinatorial therapies targeting both immune evasion and oncogenic pathways.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"42"},"PeriodicalIF":4.6,"publicationDate":"2025-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12378350/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144980416","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}
引用次数: 0
IRE1α modulates M1 oncolytic virus sensitivity via ER stress regulation in bladder cancer. IRE1α通过内质网应激调节膀胱癌中M1溶瘤病毒的敏感性。
IF 4.6
癌症耐药(英文) Pub Date : 2025-08-13 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.119
Cheng Hu, Song Wei, Wenbo Zhu, Boran Lv, Shuhao Li, Baiyu Liu, Guangmei Yan, Ying Liu
{"title":"IRE1α modulates M1 oncolytic virus sensitivity via ER stress regulation in bladder cancer.","authors":"Cheng Hu, Song Wei, Wenbo Zhu, Boran Lv, Shuhao Li, Baiyu Liu, Guangmei Yan, Ying Liu","doi":"10.20517/cdr.2025.119","DOIUrl":"10.20517/cdr.2025.119","url":null,"abstract":"<p><p><b>Aim:</b> Muscle-invasive bladder cancer (MIBC) remains lethal despite promising oncolytic virotherapy, hindered by tumor-intrinsic resistance. This study aimed to elucidate the molecular basis underlying differential sensitivity to the oncolytic M1 virus in bladder cancer. <b>Methods:</b> Bladder cancer cell lines with varying sensitivity to M1 were analyzed for endoplasmic reticulum (ER) stress responses and unfolded protein response (UPR) pathway activation. IRE1α expression was modulated using small interfering RNA and a selective inhibitor. Viral cytotoxicity, replication, and apoptosis were assessed using viability assays, immunofluorescence, electron microscopy, and immunoblotting. <i>In vivo</i> antitumor efficacy was assessed using xenografted mice. Clinical relevance was examined using patient-derived cells and survival data from The Cancer Genome Atlas. <b>Results:</b> M1 virus induced ER stress and apoptosis in sensitive cells (e.g., T24, UM-UC-3) supporting viral protein expression, whereas low-sensitivity cells like EJ showed minimal response due to limited viral replication. In moderately sensitive cells, M1 replication led to viral protein accumulation, triggering IRE1α upregulation, which in turn limited further protein buildup and apoptosis. IRE1α inhibition enhanced M1-induced ER stress, apoptotic signaling, and oncolysis without affecting viral replication capacity. <i>In vivo</i>, M1 plus STF083010 achieved greater tumor suppression than monotherapy without added toxicity. Analysis of patient-derived cells and TCGA data further revealed downregulation of IRE1α in primary tumors and its potential association with worse prognosis. <b>Conclusion:</b> IRE1α modulates M1-induced viral protein accumulation and cell death. Inhibiting IRE1α enhances ER stress and potentiates the oncolytic effect of M1 virus. Targeting IRE1α may improve M1-based virotherapy outcomes in accessible tumors.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"41"},"PeriodicalIF":4.6,"publicationDate":"2025-08-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12366421/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144980622","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}
引用次数: 0
Corosolic acid increases the therapeutic effect of cisplatin on gastric cancer by regulating Gpx4-dependent ferroptosis. 科罗索酸通过调节gpx4依赖性铁下垂提高顺铂对胃癌的治疗效果。
IF 4.6
癌症耐药(英文) Pub Date : 2025-08-07 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.94
Liubing Lin, Jian Wang, Shun Sheng, Yanting Shen, Xiaolin Liu, Rongzhong Xu, Yong Li
{"title":"Corosolic acid increases the therapeutic effect of cisplatin on gastric cancer by regulating Gpx4-dependent ferroptosis.","authors":"Liubing Lin, Jian Wang, Shun Sheng, Yanting Shen, Xiaolin Liu, Rongzhong Xu, Yong Li","doi":"10.20517/cdr.2025.94","DOIUrl":"10.20517/cdr.2025.94","url":null,"abstract":"<p><p><b>Aim:</b> Cisplatin serves as a primary chemotherapeutic agent in the treatment of gastric cancer (GC), but resistance to cisplatin-based chemotherapeutic regimens hampers its clinical application. Corosolic acid (CA), a natural triterpenoid, exhibits both anti-inflammatory and anti-cancer activities. However, the effect of CA on improving cisplatin resistance in GC remains unclear. The study primarily aimed to evaluate whether CA increases the therapeutic efficacy of cisplatin against GC and to reveal its underlying mechanism. <b>Methods:</b> Cisplatin and CA were used to treat GC cells or cisplatin-resistant AGS cells (AGS-CR), and then cell viability, apoptosis, and growth were assessed using Cell Counting Kit-8, TdT-mediated dUTP nick end labeling, and clone formation assays, respectively. Glutathione peroxidase 4 (Gpx4) expression was measured through quantitative real-time PCR and western blotting assays. <b>Results:</b> CA treatment induced a dose-dependent reduction in GC cell viability. The combination of cisplatin and CA resulted in enhanced cytotoxicity and pro-apoptotic effects compared to treatment with cisplatin alone. The effect of CA as a chemosensitizer in GC cells was damaged by a ferroptosis inhibitor, suggesting that CA decreased cisplatin chemoresistance by accelerating cancer cell ferroptosis. CA triggered cell ferroptosis by repressing Gpx4 expression in GC cells. Furthermore, elevated Gpx4 expression was significantly associated with poorer overall and disease-free survival. <b>Conclusion:</b> CA has the potential to increase cisplatin chemosensitivity in GC, and Gpx4 may represent a promising therapeutic target for its treatment.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"40"},"PeriodicalIF":4.6,"publicationDate":"2025-08-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12366429/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144980539","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}
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