癌症耐药(英文)最新文献

筛选
英文 中文
Lactylation: a novel driver of drug resistance in the tumor microenvironment. 乳酸化:肿瘤微环境中耐药的新驱动因素。
IF 4.6
癌症耐药(英文) Pub Date : 2025-08-04 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.90
Chunwei Li, Ziqiang Liu, Dezheng Kong, Zhengze Li, Lifeng Li
{"title":"Lactylation: a novel driver of drug resistance in the tumor microenvironment.","authors":"Chunwei Li, Ziqiang Liu, Dezheng Kong, Zhengze Li, Lifeng Li","doi":"10.20517/cdr.2025.90","DOIUrl":"10.20517/cdr.2025.90","url":null,"abstract":"<p><p>Lactylation, a novel lactate-derived lysine post-translational modification (PTM), has emerged as a critical epigenetic regulator driving drug resistance within the tumor microenvironment (TME). This review systematically delineates the enzymatic underpinnings of lactylation, its induction via the glycolysis-lactate axis influenced by key TME features (hypoxia, inflammation), and its multifaceted roles in promoting resistance. Specifically, lactylation orchestrates transcriptional reprogramming of resistance-associated genes (e.g., oncogenes, immune checkpoints, epithelial-mesenchymal transition factors), enhances DNA damage repair capacity (e.g., via NBS1/MRE11 lactylation), activates pro-survival autophagy, and modulates immunosuppressive signaling pathways (e.g., PI3K/AKT, NF-κB, JAK/STAT). Furthermore, it facilitates critical resistance phenotypes including immune evasion, metastasis, and angiogenesis. The review summarizes emerging therapeutic strategies targeting lactylation, such as inhibition of lactate production (LDHA/LDHB), lactate transport (MCT1/4), lactyltransferases (e.g., p300), or downstream effectors, highlighting their potential to overcome multifactorial resistance. However, elucidating the context-dependent roles, crosstalk with other PTMs, and developing specific inhibitors remain crucial for translating these insights into effective clinical interventions against resistant tumors.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"39"},"PeriodicalIF":4.6,"publicationDate":"2025-08-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12366433/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144980615","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
Effects of FAP+ cancer-associated fibroblasts on anti-PD-1 immunotherapy and CD4+ T cell polarization in gastric cancer. FAP+癌相关成纤维细胞对胃癌抗pd -1免疫治疗和CD4+ T细胞极化的影响
IF 4.6
癌症耐药(英文) Pub Date : 2025-07-29 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.97
Jing Wu, Peng-Fei Zhang, Yu Zeng, Ya-Nan Hai, Kun-Ming Zhang, Shu Dong, Ji-Chong Xu, Lan-Lin Zhang, Zhi-Xiong Wu, Hong Jiang
{"title":"Effects of FAP<sup>+</sup> cancer-associated fibroblasts on anti-PD-1 immunotherapy and CD4<sup>+</sup> T cell polarization in gastric cancer.","authors":"Jing Wu, Peng-Fei Zhang, Yu Zeng, Ya-Nan Hai, Kun-Ming Zhang, Shu Dong, Ji-Chong Xu, Lan-Lin Zhang, Zhi-Xiong Wu, Hong Jiang","doi":"10.20517/cdr.2025.97","DOIUrl":"10.20517/cdr.2025.97","url":null,"abstract":"<p><p><b>Aim:</b> The immune evasion mechanisms of gastric cancer are complex, involving various cellular dysfunctions within the tumor microenvironment. Recently, there has been growing interest in how cancer-associated fibroblasts (CAFs) contribute to tumor immune evasion. However, the precise molecular pathways through which CAFs drive immune escape in the context of gastric cancer are not yet fully elucidated. <b>Methods:</b> The abundance of FAP<sup>+</sup>CAFs in gastric cancer tissues was assessed by immunohistochemistry (IHC), and its correlation with tumor sensitivity to PD-1 monoclonal antibody therapy was analyzed. To study the effect of FAP<sup>+</sup>CAFs on naive CD4<sup>+</sup> T cell differentiation, co-culture experiments were conducted. The underlying molecular mechanisms were further investigated through western blotting and <i>in vivo</i> animal experiments. <b>Results:</b> FAP<sup>+</sup>CAFs were significantly increased in gastric cancer tissues resistant to PD-1 monoclonal antibody, and a positive correlation was found with Th2 cells. Additionally, the expression and secretion of IL-31 in FAP<sup>+</sup>CAFs cells were elevated. Mechanistically, IL-31 interacts with the IL-31R expressed on naive CD4<sup>+</sup> T cells, leading to the activation of the STAT6 signaling pathway. This cascade facilitates the differentiation of naive CD4<sup>+</sup> T cells into Th2 cells, thereby contributing to resistance against anti-PD-1 therapy in gastric cancer. <b>Conclusion:</b> FAP<sup>+</sup>CAFs may reduce sensitivity to anti-PD-1 therapy in gastric cancer by promoting Th2 polarization of naive CD4<sup>+</sup> T cells via the IL-31/STAT6 signaling pathway. Targeting this axis could offer a potential strategy to improve immunotherapy outcomes, although further validation is required.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"38"},"PeriodicalIF":4.6,"publicationDate":"2025-07-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12366491/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144980508","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
Super-enhancer inhibitors THZ2 and JQ1 reverse temozolomide resistance in glioblastoma by suppressing SE-driven SOX9 expression. 超级增强子抑制剂THZ2和JQ1通过抑制se驱动的SOX9表达逆转替莫唑胺在胶质母细胞瘤中的耐药性。
IF 4.6
癌症耐药(英文) Pub Date : 2025-07-22 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.105
Xinqi Teng, Yiming Wang, Qiang Qu, Weixin Xu, Haihui Zhuang, Yiwen Wei, Yinghuan Dai, Jian Qu
{"title":"Super-enhancer inhibitors THZ2 and JQ1 reverse temozolomide resistance in glioblastoma by suppressing SE-driven SOX9 expression.","authors":"Xinqi Teng, Yiming Wang, Qiang Qu, Weixin Xu, Haihui Zhuang, Yiwen Wei, Yinghuan Dai, Jian Qu","doi":"10.20517/cdr.2025.105","DOIUrl":"10.20517/cdr.2025.105","url":null,"abstract":"<p><p><b>Aim:</b> Glioblastoma (GBM) is the most malignant grade of glioma, characterized by high recurrence, poor prognosis, and frequent chemoresistance. There is an urgent need for alternative treatment strategies. In this study, we evaluated the effects of THZ2, a covalent inhibitor targeting the super-enhancer (SE) component CDK7, on GBM growth and chemoresistance. We also used another SE inhibitor, JQ1, to further validate the inhibitory effects of targeting SEs in GBM, thereby providing new treatment strategies for patients. <b>Methods:</b> A variety of <i>in vitro</i> and <i>in vivo</i> assays were performed to explore the anti-GBM effects of SE inhibitors. We assessed the effects of SE inhibitors in combination with temozolomide (TMZ) on GBM cells and calculated the combination index. Additionally, CUT&RUN assays were conducted to examine protein-DNA interactions. <b>Results:</b> THZ2 inhibited the proliferation, migration, and invasion of GBM cells and induced cell cycle arrest and apoptosis. Furthermore, both THZ2 and JQ1 exhibited synergistic antitumor effects when combined with TMZ in GBM cells. Notably, THZ2 reversed TMZ resistance in GBM cells by suppressing the expression of the SE-associated gene <i>SOX9</i>. We also found that <i>SOX9</i>, <i>CDK7</i>, and <i>BRD4</i> interact with histone H3K27ac. <b>Conclusion:</b> Our findings demonstrate that SE inhibitors exert antitumor effects in GBM and act synergistically with TMZ. THZ2 may enhance chemosensitivity by downregulating the SE-related gene <i>SOX9</i>, and it holds promise as a novel therapeutic agent for GBM patients.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"37"},"PeriodicalIF":4.6,"publicationDate":"2025-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12366492/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144980590","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
Decoding breast cancer treatment resistance through genetic, epigenetic, and immune-regulatory mechanisms: from molecular insights to translational perspectives. 通过遗传、表观遗传和免疫调节机制解码乳腺癌治疗耐药性:从分子视角到翻译视角。
IF 4.6
癌症耐药(英文) Pub Date : 2025-07-21 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.69
Suryendu Saha, Samikshya Mahapatra, Sinjan Khanra, Barnalee Mishra, Biswajit Swain, Diksha Malhotra, Swarnali Saha, Venketesh K Panda, Kavita Kumari, Sarmistha Jena, Sandeep Thakur, Pawan K Singh, Gopal C Kundu
{"title":"Decoding breast cancer treatment resistance through genetic, epigenetic, and immune-regulatory mechanisms: from molecular insights to translational perspectives.","authors":"Suryendu Saha, Samikshya Mahapatra, Sinjan Khanra, Barnalee Mishra, Biswajit Swain, Diksha Malhotra, Swarnali Saha, Venketesh K Panda, Kavita Kumari, Sarmistha Jena, Sandeep Thakur, Pawan K Singh, Gopal C Kundu","doi":"10.20517/cdr.2025.69","DOIUrl":"10.20517/cdr.2025.69","url":null,"abstract":"<p><p>Breast cancer continues to be the primary cause of cancer-related deaths among women globally, with increased rates of incidence and mortality, highlighting the critical need for effective treatment strategies. Recent developments have introduced a variety of treatment options that address the molecular diversity of breast cancer; nonetheless, drug resistance remains a significant barrier to achieving favorable results. This review explains the crucial role of genetic and epigenetic changes in contributing to therapeutic resistance, in addition to other factors such as increased drug efflux, enhanced DNA repair, evasion of senescence, tumor heterogeneity, the tumor microenvironment (TME), and epithelial-to-mesenchymal transition (EMT). Genetic modifications, including mutations in oncogenes and tumor suppressor genes, disrupt essential signaling pathways, facilitating resistance to chemotherapy and targeted therapies. At the same time, epigenetic modifications - like DNA methylation, alterations to histones, and dysregulation of non-coding RNAs - reprogram gene expression, supporting adaptive resistance mechanisms. These molecular abnormalities contribute to the plasticity of tumors, allowing cancer cells to evade therapeutic approaches. This review consolidates recent discoveries regarding how these genetic and epigenetic modifications affect treatment responses and resistance in breast cancer, highlighting their interaction with disease advancement. By pinpointing new drug targets, including immunotherapeutic strategies, this article seeks to shed light on the molecular underpinnings of chemoresistance, aiding in the refinement of existing treatment protocols. A more profound understanding of these mechanisms offers the potential for developing precision therapies to overcome resistance, reduce relapse rates, and improve clinical outcomes for breast cancer patients.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"36"},"PeriodicalIF":4.6,"publicationDate":"2025-07-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12366494/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144980463","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
CF10/LV overcomes acquired resistance to 5-FU/LV in colorectal cancer cells through downregulation of the c-Myc/ABCB5 axis. CF10/LV通过下调c-Myc/ABCB5轴克服结直肠癌细胞对5-FU/LV的获得性耐药。
IF 4.6
癌症耐药(英文) Pub Date : 2025-07-15 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.76
Charles Chidi Okechukwu, William H Gmeiner
{"title":"CF10/LV overcomes acquired resistance to 5-FU/LV in colorectal cancer cells through downregulation of the c-Myc/ABCB5 axis.","authors":"Charles Chidi Okechukwu, William H Gmeiner","doi":"10.20517/cdr.2025.76","DOIUrl":"10.20517/cdr.2025.76","url":null,"abstract":"<p><p><b>Aim:</b> Acquired resistance to 5-fluorouracil/leucovorin (5-FU/LV) frequently develops during treatment of metastatic colorectal (mCRC), but the causes are incompletely understood. We aim to: (i) identify the causes of 5-FU/LV resistance under physiological folate; and (ii) determine if a polymeric fluoropyrimidine (FP) CF10 remains potent to CRC cells selected for 5-FU/LV resistance. <b>Methods:</b> 5-FU/LV-resistant CRC cells were selected by repeated passaging with increasing 5-FU/LV concentrations, and resistance factors were calculated from dose-response studies. Basal and treatment-induced thymidylate synthase (TS), Myc, and ABCB5 were determined by RT-qPCR and Western blot. TS activity was determined using an <i>in situ</i> <sup>3</sup>H-release assay. DNA topoisomerase 1 cleavage complexes (Top1cc) and DNA double-strand breaks (DSBs) were determined by immunofluorescence. <b>Results:</b> Acquired resistance to 5-FU/LV with physiological folate was associated with a <1.5-fold increase in basal TS levels; however, with either 5-FU/LV or CF10/LV treatment, TS levels were elevated ~5-fold by Western blot but only ~2-fold by RT-qPCR. CF10 remained very potent to CRC cells selected for 5-FU/LV resistance, and CF10 effectively induced TS ternary complex formation and inhibited TS catalytic activity in 5-FU/LV-resistant CRC cells. c-Myc was expressed at ~4-fold higher levels in 5-FU/LV-resistant CRC cells, but Myc was barely detectable with CF10/LV treatment. The Myc-target ABCB5, which is an established factor in resistance to 5-FU and other drugs, was substantially downregulated with CF10/LV but not 5-FU/LV treatment. <b>Conclusion:</b> Acquired 5-FU/LV resistance was associated with FP-induced TS and elevated Myc and ABCB5. There is minimal cross-resistance to CF10 in 5-FU/LV-resistant CRC cells, consistent with its use in treating 5-FU/LV-resistant mCRC.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"35"},"PeriodicalIF":4.6,"publicationDate":"2025-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12367397/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144980522","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
The next frontier in antibody-drug conjugates: challenges and opportunities in cancer and autoimmune therapy. 抗体-药物结合物的下一个前沿:癌症和自身免疫治疗的挑战和机遇。
IF 4.6
癌症耐药(英文) Pub Date : 2025-07-03 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.49
Meijiang Zhou, Zhiwen Huang, Zijun Ma, Jun Chen, Shunping Lin, Xuwei Yang, Quan Gong, Zachary Braunstein, Yingying Wei, Xiaoquan Rao, Jixin Zhong
{"title":"The next frontier in antibody-drug conjugates: challenges and opportunities in cancer and autoimmune therapy.","authors":"Meijiang Zhou, Zhiwen Huang, Zijun Ma, Jun Chen, Shunping Lin, Xuwei Yang, Quan Gong, Zachary Braunstein, Yingying Wei, Xiaoquan Rao, Jixin Zhong","doi":"10.20517/cdr.2025.49","DOIUrl":"10.20517/cdr.2025.49","url":null,"abstract":"<p><p>Antibody-Drug Conjugates (ADCs) have achieved significant success in cancer therapy by combining the targeting specificity of monoclonal antibodies with cytotoxic payloads. However, the concomitant issue of drug resistance has become increasingly prominent, with primary mechanisms including alterations in target antigen expression, impaired drug transport, and inhibition of cell death pathways. ADCs have also shown emerging therapeutic potential in the treatment of autoimmune diseases; for instance, ABBV-3373 has achieved initial success in this area, yet it also faces unique challenges such as the safety of long-term administration, immunogenicity, and heterogeneity of target cells. Addressing these challenges requires multidimensional innovations, including optimizing molecular design, exploring combination therapy strategies, and introducing artificial intelligence (AI)-assisted development. These efforts aim to transition ADCs from the traditional \"targeted killing\" paradigm to intelligent and personalized precision delivery systems, thereby offering more therapeutic options for patients with cancer and autoimmune diseases.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"34"},"PeriodicalIF":4.6,"publicationDate":"2025-07-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12366496/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144980600","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
Interactions between tumor microenvironment and resistance to transarterial and systemic treatments for HCC. 肿瘤微环境与肝细胞癌经动脉和全身治疗耐药性之间的相互作用。
IF 4.6
癌症耐药(英文) Pub Date : 2025-07-02 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2024.212
Maria Stella Franzè, Francesca Saffioti, Vasileios K Mavroeidis
{"title":"Interactions between tumor microenvironment and resistance to transarterial and systemic treatments for HCC.","authors":"Maria Stella Franzè, Francesca Saffioti, Vasileios K Mavroeidis","doi":"10.20517/cdr.2024.212","DOIUrl":"10.20517/cdr.2024.212","url":null,"abstract":"<p><p>Hepatocellular carcinoma (HCC) is a malignant tumor originating from hepatocytes, often developing against a backdrop of chronic inflammation and liver fibrosis. The primary risk factor for HCC is cirrhosis, and early detection is crucial for improving outcomes. Despite advances in treatment, the prognosis remains poor, with a 5-year survival rate of approximately 15%-38%. Growing evidence highlights the critical role of the tumor microenvironment (TME) in modulating tumor initiation, growth, progression, and, in some cases, suppression. The TME is a complex ecosystem composed of immune cells, cancer-associated fibroblasts, extracellular matrix components, and other factors such as growth factors and cytokines. By shaping tumor cell behavior, the TME facilitates immune evasion and contributes to resistance to treatment. Tumor-associated immune cells, including regulatory T cells, myeloid-derived suppressor cells, and tumor-associated macrophages, contribute to immune suppression and progression. On the other hand, immune activation via immune checkpoint inhibition has shown promise in improving outcomes, especially when combined with other treatments such as transarterial chemoembolization (TACE), selective internal radiation therapy (SIRT), and systemic therapies. Studies have demonstrated the potential of targeting the TME to enhance treatment efficacy, with immune modulation emerging as a key therapeutic strategy. This review explores the complex interactions within the TME in HCC, highlighting its role in therapy resistance and immune evasion. It also discusses current therapeutic approaches to target the TME to improve clinical outcomes in HCC patients.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"33"},"PeriodicalIF":4.6,"publicationDate":"2025-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12366493/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144980617","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
Modeling methods of different tumor organoids and their application in tumor drug resistance research. 不同肿瘤类器官建模方法及其在肿瘤耐药研究中的应用。
IF 4.6
癌症耐药(英文) Pub Date : 2025-07-01 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.34
Chengming Yang, Lushan Yang, Yuchen Feng, Xingyi Song, Shu Bai, Sheng Zhang, Mingjuan Sun
{"title":"Modeling methods of different tumor organoids and their application in tumor drug resistance research.","authors":"Chengming Yang, Lushan Yang, Yuchen Feng, Xingyi Song, Shu Bai, Sheng Zhang, Mingjuan Sun","doi":"10.20517/cdr.2025.34","DOIUrl":"10.20517/cdr.2025.34","url":null,"abstract":"<p><p>Tumor organoids were modeled <i>in vitro</i> to mimic <i>in vivo</i> culture conditions, allowing tumor-derived tissue cells or isolated and purified tumor stem cells to self-assemble into 3D preclinical models that are similar to tissues and organs <i>in vivo</i>. Compared with traditional models, tumor organoids not only resemble parental tumors in histology and genomics, capturing their heterogeneity and drug response, but also provide an efficient platform for long-term culture, maintaining genetic stability and enabling gene manipulation. Therefore, tumor organoids have unique advantages in cancer drug resistance research. The paper covers: (1) Modeling methods of epithelial and non-epithelial tumor organoids, with special emphasis on the modeling of drug-resistant organoids; (2) Their use in drug resistance research, split into i. Therapeutic exploration (drug testing and screening) and ii. Mechanism investigation (use drug-resistant organoids to study drug resistance), including methods and findings from various teams.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"32"},"PeriodicalIF":4.6,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12366422/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144980627","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
Integrated multi-omics profiling of immune microenvironment and drug resistance signatures for precision prognosis in prostate cancer. 前列腺癌免疫微环境和耐药特征的综合多组学分析用于精确预后。
IF 4.6
癌症耐药(英文) Pub Date : 2025-06-25 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.47
Chao Li, Longxiang Wu, Bowen Zhong, Yu Gan, Lei Zhou, Shuo Tan, Weibin Hou, Kun Yao, Bingzhi Wang, Zhenyu Ou, Shengwang Zhang, Wei Xiong
{"title":"Integrated multi-omics profiling of immune microenvironment and drug resistance signatures for precision prognosis in prostate cancer.","authors":"Chao Li, Longxiang Wu, Bowen Zhong, Yu Gan, Lei Zhou, Shuo Tan, Weibin Hou, Kun Yao, Bingzhi Wang, Zhenyu Ou, Shengwang Zhang, Wei Xiong","doi":"10.20517/cdr.2025.47","DOIUrl":"10.20517/cdr.2025.47","url":null,"abstract":"<p><p><b>Introduction:</b> Prostate cancer (PCa) continues to be a significant cause of mortality among men, with treatment resistance often influenced by the complexity of the tumor microenvironment (TME). This study aims to develop an immune-centric prognostic model that correlates TME dynamics, genomic instability, and the heterogeneity of drug resistance in PCa. <b>Methods:</b> Multi-omics data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases were integrated, encompassing transcriptomic profiles of 554 TCGA-PRAD samples and 329 external validation samples. Immune cell infiltration was assessed using CIBERSORT and ESTIMATE. Weighted gene co-expression network analysis (WGCNA) was employed to identify immune-related modules. Single-cell RNA sequencing (ScRNA-seq) of 835 PCa cells uncovered subtype-specific resistance patterns. Prognostic models were constructed using least absolute shrinkage and selection operator (LASSO) regression and subsequently validated experimentally in PCa cell lines. <b>Results:</b> Two immune subtypes were identified: high-risk subgroups displayed TP53 mutations, increased tumor mutation burden (TMB), and enriched energy metabolism pathways. ScRNA-seq delineated three PCa cell clusters, with high-risk subtypes being sensitive to bendamustine/dacomitinib and resistant to apalutamide/neratinib. A 10-gene prognostic model (e.g., MUC5B, TREM1) categorized patients into high/low-risk groups with distinct survival outcomes (log-rank <i>P</i> < 0.0001). Validation in external datasets confirmed the robust predictive accuracy (AUC: 0.854-0.889). Experimental assays verified subtype-specific drug responses and dysregulation of key model genes. <b>Discussion:</b> This study establishes a TME-driven prognostic framework that connects immune heterogeneity, genomic instability, and therapeutic resistance in PCa. By pinpointing metabolic dependencies and subtype-specific vulnerabilities, our findings provide actionable strategies to circumvent treatment failure, such as targeting energy metabolism or tailoring therapies based on resistance signatures.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"31"},"PeriodicalIF":4.6,"publicationDate":"2025-06-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12366425/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144980457","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
MicroRNA-mediated autophagy regulation in thyroid cancer drug resistance. 微rna介导的甲状腺癌耐药自噬调控。
IF 4.6
癌症耐药(英文) Pub Date : 2025-06-18 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.73
Dongye Huang, Qianwen Liu, Chang Liu, Jingna Cao, Senmin Zhang, Huijiao Cao, Wenkuan Chen
{"title":"MicroRNA-mediated autophagy regulation in thyroid cancer drug resistance.","authors":"Dongye Huang, Qianwen Liu, Chang Liu, Jingna Cao, Senmin Zhang, Huijiao Cao, Wenkuan Chen","doi":"10.20517/cdr.2025.73","DOIUrl":"10.20517/cdr.2025.73","url":null,"abstract":"<p><p>Thyroid cancer, particularly papillary thyroid cancer (PTC), represents the most prevalent endocrine malignancy. Despite advancements in therapeutic strategies, drug resistance significantly hampers clinical outcomes. Autophagy, an evolutionarily conserved cellular degradation pathway, acts paradoxically in thyroid cancer by promoting either tumor cell survival or cell death, thus influencing therapeutic resistance. Increasing evidence highlights microRNAs (miRNAs), small non-coding RNAs, as critical regulators of autophagy through precise modulation of autophagy-related genes (ATGs) and signaling pathways. miRNA-mediated autophagy can either enhance chemotherapeutic efficacy or facilitate resistance, depending on the cellular context and miRNA targets. This review summarizes recent insights into miRNA-autophagy interactions underlying drug resistance in thyroid cancer, emphasizing key miRNAs, including miR-125b, miR-144, miR-30d, and miR-9-5p. Understanding the complex regulatory networks connecting miRNAs and autophagy provides promising avenues for developing novel therapeutic strategies to overcome resistance in refractory thyroid cancer.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"30"},"PeriodicalIF":4.6,"publicationDate":"2025-06-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12366428/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144980558","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