首页 > 最新文献

癌症耐药(英文)最新文献

英文 中文
FGFR1 overexpression promotes resistance to PI3K inhibitor alpelisib in luminal breast cancer cells through receptor tyrosine kinase signaling-mediated activation of the estrogen receptor. FGFR1过表达通过受体酪氨酸激酶信号介导的雌激素受体激活促进腔内乳腺癌细胞对PI3K抑制剂alpelisib的抗性。
IF 4.6 Q1 ONCOLOGY Pub Date : 2025-05-28 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2024.181
Yujie Shi, Lexia Chen, Qiong Cheng, Peijia Niu, Yahan Weng, Xiaohe Yang

Aim: Resistance to PI3K inhibitor alpelisib is an emerging challenge in breast cancer treatment. FGFR1 is frequently amplified in breast cancer. We investigated FGFR1 overexpression-mediated alpelisib resistance and its mechanism. Methods: CCK-8, colony formation, and cell cycle assays assessed FGFR1 overexpression-induced alpelisib resistance in MCF-7 and T47D cells. FGFR1 siRNA knockdown validated FGFR1's role. Akt, Erk, and ER signaling were analyzed by Western blot. Synergistic effects of alpelisib with AZD4547 and fulvestrant were evaluated using the combination index. Results: FGFR1 overexpression conferred alpelisib resistance in MCF-7 and T47D cells, evidenced by increased viability, colony formation, and S-phase accumulation post alpelisib treatment. Knockdown of FGFR1 reverse alpelisib resistance in FGFR1 overexpressing MCF-7 and T47D cells. Resistance correlated with sustained activation of Akt and Erk1/2 pathways (p-Akt, p-Erk1/2, p-S6K, p-Rb) and attenuated suppression of ERα phosphorylation (S118/S167), highlighting RTK-ER crosstalk. Combining alpelisib with AZD4547 synergistically inhibited growth and suppressed both RTK signaling and ERα phosphorylation. While alpelisib-fulvestrant was effective, adding AZD4547 further enhanced inhibition, supporting triple therapy to overcome resistance. Conclusion: Our findings establish FGFR1 as a key mediator of alpelisib resistance in ER+ breast cancer. Combining FGFR1 inhibitors with alpelisib-based therapies offers a viable approach for FGFR1-overexpressing tumors.

目的:对PI3K抑制剂alpelisib的耐药是乳腺癌治疗中的一个新挑战。FGFR1在乳腺癌中经常被扩增。我们研究了FGFR1过表达介导的alpelisib耐药及其机制。方法:CCK-8、菌落形成和细胞周期检测评估MCF-7和T47D细胞中FGFR1过表达诱导的alpelisib抗性。FGFR1 siRNA敲低证实了FGFR1的作用。Western blot分析Akt、Erk和ER信号。采用联合指数评价alpelisib与AZD4547和氟维司汀的协同效应。结果:在MCF-7和T47D细胞中,FGFR1过表达赋予了alpelisib抗性,证明了alpelisib处理后细胞活力、菌落形成和s期积累的增加。FGFR1敲低可逆转MCF-7和T47D细胞中FGFR1过表达的alpelisib耐药性。耐药与Akt和Erk1/2通路(p-Akt, p-Erk1/2, p-S6K, p-Rb)的持续激活和ERα磷酸化抑制减弱(S118/S167)相关,突出RTK-ER串扰。alpelisib与AZD4547合用可协同抑制生长,抑制RTK信号和ERα磷酸化。虽然alpelisib-fulvestrant是有效的,但添加AZD4547进一步增强了抑制作用,支持三联疗法克服耐药性。结论:我们的研究结果证实FGFR1是ER+乳腺癌中alpelisib耐药的关键介质。结合FGFR1抑制剂和基于alpelisib的治疗为FGFR1过表达肿瘤提供了一种可行的方法。
{"title":"FGFR1 overexpression promotes resistance to PI3K inhibitor alpelisib in luminal breast cancer cells through receptor tyrosine kinase signaling-mediated activation of the estrogen receptor.","authors":"Yujie Shi, Lexia Chen, Qiong Cheng, Peijia Niu, Yahan Weng, Xiaohe Yang","doi":"10.20517/cdr.2024.181","DOIUrl":"10.20517/cdr.2024.181","url":null,"abstract":"<p><p><b>Aim:</b> Resistance to PI3K inhibitor alpelisib is an emerging challenge in breast cancer treatment. FGFR1 is frequently amplified in breast cancer. We investigated FGFR1 overexpression-mediated alpelisib resistance and its mechanism. <b>Methods:</b> CCK-8, colony formation, and cell cycle assays assessed FGFR1 overexpression-induced alpelisib resistance in MCF-7 and T47D cells. FGFR1 siRNA knockdown validated FGFR1's role. Akt, Erk, and ER signaling were analyzed by Western blot. Synergistic effects of alpelisib with AZD4547 and fulvestrant were evaluated using the combination index. <b>Results:</b> FGFR1 overexpression conferred alpelisib resistance in MCF-7 and T47D cells, evidenced by increased viability, colony formation, and S-phase accumulation post alpelisib treatment. Knockdown of FGFR1 reverse alpelisib resistance in FGFR1 overexpressing MCF-7 and T47D cells. Resistance correlated with sustained activation of Akt and Erk1/2 pathways (p-Akt, p-Erk1/2, p-S6K, p-Rb) and attenuated suppression of ERα phosphorylation (S118/S167), highlighting RTK-ER crosstalk. Combining alpelisib with AZD4547 synergistically inhibited growth and suppressed both RTK signaling and ERα phosphorylation. While alpelisib-fulvestrant was effective, adding AZD4547 further enhanced inhibition, supporting triple therapy to overcome resistance. <b>Conclusion:</b> Our findings establish FGFR1 as a key mediator of alpelisib resistance in ER+ breast cancer. Combining FGFR1 inhibitors with alpelisib-based therapies offers a viable approach for FGFR1-overexpressing tumors.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"24"},"PeriodicalIF":4.6,"publicationDate":"2025-05-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12159599/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144287384","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
Advances in the application of patient-derived xenograft models in acute leukemia resistance. 患者源性异种移植模型在急性白血病耐药研究中的应用进展。
IF 4.6 Q1 ONCOLOGY Pub Date : 2025-05-28 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.18
Ronghao Qin, Yuxing Liang, Fuling Zhou

Acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) are genetically heterogeneous malignancies of hematopoietic stem cells, characterized by complex mutations and a high risk of drug resistance and relapse. Patient-derived xenograft (PDX) models are dynamic entities transplanted with leukemia stem cells (LSCs), retaining patients' biological and genetic characteristics. By elucidating LSCs, clonal dynamics, and microenvironment interaction, PDXs facilitate the preclinical evaluation of therapy sensitivity, including immunotherapies, epigenetic therapies, and other agents targeting mutated proteins or apoptosis. The application of PDXs has provided translational evidence for various studies with reliable clinical relevance. Additionally, conventional PDXs remain a robust tool in identifying drug resistance compared with other models, and their potential is further unleashed when examined in large cohorts or combined with novel technologies, which not only enhances our understanding of acute leukemia biology but also enables the discovery and identification of novel biomarkers. In this review, we present the application of PDX models for acute leukemia resistance, including mechanism investigation, therapy evaluation, and associated challenges.

急性髓性白血病(AML)和急性淋巴细胞白血病(ALL)是造血干细胞的遗传异质性恶性肿瘤,具有复杂突变和高耐药和复发风险的特点。患者来源的异种移植(PDX)模型是移植白血病干细胞(LSCs)的动态实体,保留了患者的生物学和遗传特征。通过阐明LSCs、克隆动力学和微环境相互作用,PDXs促进了治疗敏感性的临床前评估,包括免疫疗法、表观遗传疗法和其他靶向突变蛋白或细胞凋亡的药物。pdx的应用为各种具有可靠临床相关性的研究提供了翻译证据。此外,与其他模型相比,传统的pdx仍然是鉴定耐药的强大工具,当在大型队列中进行检测或与新技术相结合时,它们的潜力进一步释放,这不仅增强了我们对急性白血病生物学的理解,而且还使新的生物标志物的发现和鉴定成为可能。在这篇综述中,我们介绍了PDX模型在急性白血病耐药中的应用,包括机制研究、治疗评估和相关挑战。
{"title":"Advances in the application of patient-derived xenograft models in acute leukemia resistance.","authors":"Ronghao Qin, Yuxing Liang, Fuling Zhou","doi":"10.20517/cdr.2025.18","DOIUrl":"10.20517/cdr.2025.18","url":null,"abstract":"<p><p>Acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) are genetically heterogeneous malignancies of hematopoietic stem cells, characterized by complex mutations and a high risk of drug resistance and relapse. Patient-derived xenograft (PDX) models are dynamic entities transplanted with leukemia stem cells (LSCs), retaining patients' biological and genetic characteristics. By elucidating LSCs, clonal dynamics, and microenvironment interaction, PDXs facilitate the preclinical evaluation of therapy sensitivity, including immunotherapies, epigenetic therapies, and other agents targeting mutated proteins or apoptosis. The application of PDXs has provided translational evidence for various studies with reliable clinical relevance. Additionally, conventional PDXs remain a robust tool in identifying drug resistance compared with other models, and their potential is further unleashed when examined in large cohorts or combined with novel technologies, which not only enhances our understanding of acute leukemia biology but also enables the discovery and identification of novel biomarkers. In this review, we present the application of PDX models for acute leukemia resistance, including mechanism investigation, therapy evaluation, and associated challenges.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"23"},"PeriodicalIF":4.6,"publicationDate":"2025-05-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12159603/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144287383","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
Unveiling MAGEA3: a novel predictive biomarker for bevacizumab resistance in colorectal cancer. 揭示MAGEA3:结直肠癌贝伐单抗耐药的新型预测性生物标志物
IF 4.6 Q1 ONCOLOGY Pub Date : 2025-04-28 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.35
Juncheng Su, Jiahui Wang, Weilin Chen, Yingjie Xu, Wen Yang, Weiwei Liu, Zheng Wang, Masha Huang

Aim: Bevacizumab has long been a cornerstone in the treatment of colorectal cancer (CRC), serving as a fundamental antiangiogenic therapeutic option. However, a significant proportion of patients exhibit insensitivity to bevacizumab, and no reliable biomarker has been established to predict treatment efficacy. Notably, while many angiogenic factors in tumors have been extensively studied, they have failed to consistently demonstrate reliable predictive value for patient survival outcomes in CRC. This study is designed to screen tumor biomarkers with predictive value for bevacizumab resistance in CRC. Methods: Online CRC databases with bevacizumab treatment were downloaded from the GEO datasets along with the TCGA database, which were then analyzed to generate genes overexpressed in bevacizumab non-responders. In vitro experiments using colorectal cancer cell lines were then performed to explore the underlying mechanism of the candidate gene that impacts bevacizumab efficacy. Finally, clinical samples of CRC were collected to validate the predictive effect of the candidate gene on bevacizumab efficacy. Results: We conducted comprehensive analyses of CRC patient datasets, identifying MAGEA3 as a pivotal gene that is not only highly upregulated in bevacizumab-resistant primary CRC but also strongly associated with poor overall survival prognosis. Our in vitro experiments revealed a novel mechanistic insight: MAGEA3 specifically inhibits the expression and secretion of VEGF through the mTOR signaling pathway in colorectal cancer cells, while exhibiting minimal impact on other key angiogenic factors such as PDGF, FGF, and ANGPT2. This selective regulation of VEGF provides a molecular basis for MAGEA3's role in bevacizumab resistance. Furthermore, we discovered that MAGEA3 significantly impairs mitochondrial function in cancer cells, suggesting an additional layer of complexity in its oncogenic role. Clinically, our findings demonstrated that high baseline levels of MAGEA3 in CRC patients were strongly associated with worse progression-free survival (PFS) following bevacizumab treatment. Conclusion: Collectively, these findings position MAGEA3 as a promising predictive biomarker for bevacizumab resistance in CRC, offering a potential solution to the longstanding challenge of treatment stratification.

目的:贝伐单抗长期以来一直是结直肠癌(CRC)治疗的基石,作为一种基本的抗血管生成治疗选择。然而,相当比例的患者对贝伐单抗不敏感,并且没有建立可靠的生物标志物来预测治疗效果。值得注意的是,虽然肿瘤中的许多血管生成因子已被广泛研究,但它们未能始终如一地证明对CRC患者生存结果的可靠预测价值。本研究旨在筛选CRC中具有贝伐单抗耐药预测价值的肿瘤生物标志物。方法:从GEO数据集和TCGA数据库中下载贝伐珠单抗治疗的在线CRC数据库,然后对其进行分析,以生成贝伐珠单抗无应答者的过表达基因。然后使用结直肠癌细胞系进行体外实验,以探索影响贝伐单抗疗效的候选基因的潜在机制。最后,收集CRC临床样本,验证候选基因对贝伐单抗疗效的预测作用。结果:我们对结直肠癌患者数据集进行了全面分析,发现MAGEA3是一个关键基因,不仅在贝伐单抗耐药的原发性结直肠癌中高度上调,而且与不良的总生存预后密切相关。我们的体外实验揭示了一个新的机制:MAGEA3通过mTOR信号通路特异性抑制结直肠癌细胞中VEGF的表达和分泌,而对其他关键血管生成因子如PDGF、FGF和ANGPT2的影响很小。这种对VEGF的选择性调控为MAGEA3在贝伐单抗耐药中的作用提供了分子基础。此外,我们发现MAGEA3显著损害癌细胞中的线粒体功能,这表明其致癌作用的另一层复杂性。临床上,我们的研究结果表明,在贝伐单抗治疗后,CRC患者中高基线水平的MAGEA3与较差的无进展生存期(PFS)密切相关。结论:总的来说,这些发现将MAGEA3定位为结直肠癌贝伐单抗耐药的有希望的预测性生物标志物,为长期存在的治疗分层挑战提供了潜在的解决方案。
{"title":"Unveiling MAGEA3: a novel predictive biomarker for bevacizumab resistance in colorectal cancer.","authors":"Juncheng Su, Jiahui Wang, Weilin Chen, Yingjie Xu, Wen Yang, Weiwei Liu, Zheng Wang, Masha Huang","doi":"10.20517/cdr.2025.35","DOIUrl":"https://doi.org/10.20517/cdr.2025.35","url":null,"abstract":"<p><p><b>Aim:</b> Bevacizumab has long been a cornerstone in the treatment of colorectal cancer (CRC), serving as a fundamental antiangiogenic therapeutic option. However, a significant proportion of patients exhibit insensitivity to bevacizumab, and no reliable biomarker has been established to predict treatment efficacy. Notably, while many angiogenic factors in tumors have been extensively studied, they have failed to consistently demonstrate reliable predictive value for patient survival outcomes in CRC. This study is designed to screen tumor biomarkers with predictive value for bevacizumab resistance in CRC. <b>Methods:</b> Online CRC databases with bevacizumab treatment were downloaded from the GEO datasets along with the TCGA database, which were then analyzed to generate genes overexpressed in bevacizumab non-responders. In vitro experiments using colorectal cancer cell lines were then performed to explore the underlying mechanism of the candidate gene that impacts bevacizumab efficacy. Finally, clinical samples of CRC were collected to validate the predictive effect of the candidate gene on bevacizumab efficacy. <b>Results:</b> We conducted comprehensive analyses of CRC patient datasets, identifying MAGEA3 as a pivotal gene that is not only highly upregulated in bevacizumab-resistant primary CRC but also strongly associated with poor overall survival prognosis. Our in vitro experiments revealed a novel mechanistic insight: MAGEA3 specifically inhibits the expression and secretion of VEGF through the mTOR signaling pathway in colorectal cancer cells, while exhibiting minimal impact on other key angiogenic factors such as PDGF, FGF, and ANGPT2. This selective regulation of VEGF provides a molecular basis for MAGEA3's role in bevacizumab resistance. Furthermore, we discovered that MAGEA3 significantly impairs mitochondrial function in cancer cells, suggesting an additional layer of complexity in its oncogenic role. Clinically, our findings demonstrated that high baseline levels of MAGEA3 in CRC patients were strongly associated with worse progression-free survival (PFS) following bevacizumab treatment. <b>Conclusion:</b> Collectively, these findings position MAGEA3 as a promising predictive biomarker for bevacizumab resistance in CRC, offering a potential solution to the longstanding challenge of treatment stratification.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"22"},"PeriodicalIF":4.6,"publicationDate":"2025-04-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12059477/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144021765","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
Tumor microenvironment-driven resistance to immunotherapy in non-small cell lung cancer: strategies for Cold-to-Hot tumor transformation. 非小细胞肺癌肿瘤微环境驱动免疫治疗耐药:冷-热肿瘤转化策略
IF 4.6 Q1 ONCOLOGY Pub Date : 2025-04-24 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.14
Jinglu Yu, Xiaoni Kong, Yu Feng

Non-small cell lung cancer (NSCLC) represents a formidable challenge in oncology due to its molecular heterogeneity and the dynamic suppressive nature of its tumor microenvironment (TME). Despite the transformative impact of immune checkpoint inhibitors (ICIs) on cancer therapy, the majority of NSCLC patients experience resistance, necessitating novel approaches to overcome immune evasion. This review highlights shared and subtype-specific mechanisms of immune resistance within the TME, including metabolic reprogramming, immune cell dysfunction, and physical barriers. Beyond well-characterized components such as regulatory T cells, tumor-associated macrophages, and myeloid-derived suppressor cells, emerging players - neutrophil extracellular traps, tertiary lymphoid structures, and exosomal signaling networks - underscore the TME's complexity and adaptability. A multi-dimensional framework is proposed to transform cold, immune-excluded tumors into hot, immune-reactive ones. Key strategies include enhancing immune infiltration, modulating immunosuppressive networks, and activating dormant immune pathways. Cutting-edge technologies, such as single-cell sequencing, spatial transcriptomics, and nanomedicine, are identified as pivotal tools for decoding TME heterogeneity and personalizing therapeutic interventions. By bridging mechanistic insights with translational innovations, this review advocates for integrative approaches that combine ICIs with metabolic modulators, vascular normalizers, and emerging therapies such as STING agonists and tumor vaccines. The synergistic potential of these strategies is poised to overcome resistance and achieve durable antitumor immunity. Ultimately, this vision underscores the importance of interdisciplinary collaboration and real-time TME profiling in refining precision oncology for NSCLC, offering a blueprint for extending these advances to other malignancies.

非小细胞肺癌(NSCLC)由于其分子异质性和肿瘤微环境(TME)的动态抑制特性,在肿瘤学领域是一个巨大的挑战。尽管免疫检查点抑制剂(ICIs)对癌症治疗具有变革性影响,但大多数非小细胞肺癌患者都存在耐药性,因此需要新的方法来克服免疫逃避。这篇综述强调了TME中共有的和亚型特异性的免疫抵抗机制,包括代谢重编程、免疫细胞功能障碍和物理障碍。除了特征明确的成分,如调节性T细胞、肿瘤相关巨噬细胞和髓源性抑制细胞外,中性粒细胞胞外陷阱、三级淋巴结构和外泌体信号网络等新兴成分强调了TME的复杂性和适应性。提出了一种多维框架,将冷的、免疫排斥的肿瘤转化为热的、免疫反应的肿瘤。关键策略包括增强免疫浸润、调节免疫抑制网络和激活休眠免疫途径。尖端技术,如单细胞测序、空间转录组学和纳米医学,被认为是解码TME异质性和个性化治疗干预的关键工具。通过将机制见解与转化创新联系起来,本综述提倡将ICIs与代谢调节剂、血管正常化剂和新兴疗法(如STING激动剂和肿瘤疫苗)结合起来的综合方法。这些策略的协同潜力有望克服耐药性并实现持久的抗肿瘤免疫。最终,这一愿景强调了跨学科合作和实时TME分析在改进非小细胞肺癌精确肿瘤学中的重要性,为将这些进展扩展到其他恶性肿瘤提供了蓝图。
{"title":"Tumor microenvironment-driven resistance to immunotherapy in non-small cell lung cancer: strategies for Cold-to-Hot tumor transformation.","authors":"Jinglu Yu, Xiaoni Kong, Yu Feng","doi":"10.20517/cdr.2025.14","DOIUrl":"https://doi.org/10.20517/cdr.2025.14","url":null,"abstract":"<p><p>Non-small cell lung cancer (NSCLC) represents a formidable challenge in oncology due to its molecular heterogeneity and the dynamic suppressive nature of its tumor microenvironment (TME). Despite the transformative impact of immune checkpoint inhibitors (ICIs) on cancer therapy, the majority of NSCLC patients experience resistance, necessitating novel approaches to overcome immune evasion. This review highlights shared and subtype-specific mechanisms of immune resistance within the TME, including metabolic reprogramming, immune cell dysfunction, and physical barriers. Beyond well-characterized components such as regulatory T cells, tumor-associated macrophages, and myeloid-derived suppressor cells, emerging players - neutrophil extracellular traps, tertiary lymphoid structures, and exosomal signaling networks - underscore the TME's complexity and adaptability. A multi-dimensional framework is proposed to transform cold, immune-excluded tumors into hot, immune-reactive ones. Key strategies include enhancing immune infiltration, modulating immunosuppressive networks, and activating dormant immune pathways. Cutting-edge technologies, such as single-cell sequencing, spatial transcriptomics, and nanomedicine, are identified as pivotal tools for decoding TME heterogeneity and personalizing therapeutic interventions. By bridging mechanistic insights with translational innovations, this review advocates for integrative approaches that combine ICIs with metabolic modulators, vascular normalizers, and emerging therapies such as STING agonists and tumor vaccines. The synergistic potential of these strategies is poised to overcome resistance and achieve durable antitumor immunity. Ultimately, this vision underscores the importance of interdisciplinary collaboration and real-time TME profiling in refining precision oncology for NSCLC, offering a blueprint for extending these advances to other malignancies.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"21"},"PeriodicalIF":4.6,"publicationDate":"2025-04-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12059482/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144060509","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
Ivosidenib enhances cisplatin sensitivity in ovarian cancer by reducing cancer cell stemness. Ivosidenib通过降低癌细胞干细胞性增强卵巢癌顺铂敏感性。
IF 4.6 Q1 ONCOLOGY Pub Date : 2025-04-24 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.51
Mengqing Chen, Lin Huang, Simei Zhao, Mengna Zhu, Si Sun, Wenhan Li, Jing Cai, Minggang Peng, Yiping Wen, Zehua Wang

Aim: Cancer stem cells (CSCs) are pivotal in mediating platinum resistance in ovarian cancer. This study aimed to screen compounds sensitizing CSCs to cisplatin by using a small molecule inhibitor library. Methods: A library of 105 common drugs was screened in ovarian CSC model SK-3rd and ovarian cancer platinum-resistant cell model SKDDP to identify those that could enhance sensitivity to cisplatin by MTT assay. The antitumor effect was assessed in ovarian cancer cells using the MTT assay, colony formation assay, and apoptosis assay. The impact on cancer cell stemness was evaluated using qPCR and Sphere-forming assays. Finally, the effect of the combination regimen was evaluated in patient-derived organoids (PDOs) under different treatments by the CellTiter-Glo Luminescence Assay. Results: The results of the initial screening on SK-3rd identified five candidate compounds. Rescreening on SKDDP showed that Ivosidenib was the most effective in sensitizing cisplatin. MTT, colony formation, and apoptosis assays demonstrated that Ivosidenib enhanced the sensitivity to cisplatin, inhibited proliferation, and induced apoptosis in ovarian cancer cells, including SK-3rd and SKDDP. Furthermore, Ivosidenib lowered stemness marker expression and countered CSC enrichment caused by platinum-based chemotherapy in ovarian cancer cells. Finally, the synergistic effect of this combination was also confirmed in three ovarian cancer PDOs. Conclusion: Ivosidenib may increase cisplatin sensitivity in ovarian cancer cells by decreasing their stemness, providing a potential therapeutic method for ovarian cancer patients.

目的:肿瘤干细胞(CSCs)在介导卵巢癌铂耐药中起关键作用。本研究旨在利用小分子抑制剂文库筛选使CSCs对顺铂敏感的化合物。方法:从卵巢CSC模型sk -3和卵巢癌耐铂细胞模型SKDDP中筛选出105种常用药物,采用MTT法筛选能增强顺铂敏感性的药物。使用MTT试验、菌落形成试验和细胞凋亡试验评估卵巢癌细胞的抗肿瘤作用。使用qPCR和球形成实验评估对癌细胞干细胞的影响。最后,通过细胞滴度-荧光荧光法(CellTiter-Glo Luminescence Assay)评估联合方案在不同治疗下患者源性类器官(PDOs)中的效果。结果:sk -3初步筛选筛选出5个候选化合物。SKDDP再筛选显示Ivosidenib对顺铂增敏效果最好。MTT、集落形成和细胞凋亡实验表明,Ivosidenib增强了卵巢癌细胞(包括sk -3和SKDDP)对顺铂的敏感性,抑制了增殖,并诱导了细胞凋亡。此外,Ivosidenib降低了卵巢癌细胞中干细胞标志物的表达,并对抗了铂基化疗引起的CSC富集。最后,在3例卵巢癌pdo中也证实了该组合的协同作用。结论:Ivosidenib可能通过降低卵巢癌细胞的干性而增加卵巢癌细胞对顺铂的敏感性,为卵巢癌患者提供了一种潜在的治疗方法。
{"title":"Ivosidenib enhances cisplatin sensitivity in ovarian cancer by reducing cancer cell stemness.","authors":"Mengqing Chen, Lin Huang, Simei Zhao, Mengna Zhu, Si Sun, Wenhan Li, Jing Cai, Minggang Peng, Yiping Wen, Zehua Wang","doi":"10.20517/cdr.2025.51","DOIUrl":"https://doi.org/10.20517/cdr.2025.51","url":null,"abstract":"<p><p><b>Aim</b>: Cancer stem cells (CSCs) are pivotal in mediating platinum resistance in ovarian cancer. This study aimed to screen compounds sensitizing CSCs to cisplatin by using a small molecule inhibitor library. <b>Methods</b>: A library of 105 common drugs was screened in ovarian CSC model SK-3rd and ovarian cancer platinum-resistant cell model SKDDP to identify those that could enhance sensitivity to cisplatin by MTT assay. The antitumor effect was assessed in ovarian cancer cells using the MTT assay, colony formation assay, and apoptosis assay. The impact on cancer cell stemness was evaluated using qPCR and Sphere-forming assays. Finally, the effect of the combination regimen was evaluated in patient-derived organoids (PDOs) under different treatments by the CellTiter-Glo Luminescence Assay. <b>Results</b>: The results of the initial screening on SK-3rd identified five candidate compounds. Rescreening on SKDDP showed that Ivosidenib was the most effective in sensitizing cisplatin. MTT, colony formation, and apoptosis assays demonstrated that Ivosidenib enhanced the sensitivity to cisplatin, inhibited proliferation, and induced apoptosis in ovarian cancer cells, including SK-3rd and SKDDP. Furthermore, Ivosidenib lowered stemness marker expression and countered CSC enrichment caused by platinum-based chemotherapy in ovarian cancer cells. Finally, the synergistic effect of this combination was also confirmed in three ovarian cancer PDOs. <b>Conclusion</b>: Ivosidenib may increase cisplatin sensitivity in ovarian cancer cells by decreasing their stemness, providing a potential therapeutic method for ovarian cancer patients.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"20"},"PeriodicalIF":4.6,"publicationDate":"2025-04-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12059478/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144036543","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 genetic architecture of bone metastases: unveiling the role of epigenetic and genetic modifications in drug resistance. 骨转移的遗传结构:揭示表观遗传和遗传修饰在耐药性中的作用。
IF 4.6 Q1 ONCOLOGY Pub Date : 2025-04-22 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.28
Ahmad Dawalibi, Mohamad Bakir, Khalid S Mohammad

Bone metastases represent frequent and severe complications in various cancers, notably impacting prognosis and quality of life. This review article delves into the genetic and epigenetic mechanisms underpinning drug resistance in bone metastases, a key challenge in effective cancer treatment. The development of drug resistance in cancer can manifest as either intrinsic or acquired, with genetic heterogeneity playing a pivotal role. Intrinsic resistance is often due to pre-existing mutations, while acquired resistance evolves through genetic and epigenetic alterations during treatment. These alterations include mutations in driver genes like TP53 and RB1, epigenetic modifications such as DNA methylation and histone changes, and pathway alterations, notably involving RANK-RANKL signaling and the PI3K/AKT/mTOR cascade. Recent studies underline the significance of the tumor microenvironment in fostering drug resistance, with components such as cancer-associated fibroblasts and hypoxia playing crucial roles. The interactions between metastatic cancer cells and the bone microenvironment facilitate survival and the proliferation of drug-resistant clones. This review highlights the necessity of understanding these complex interactions to develop targeted therapies that can overcome resistance and improve treatment outcomes. Current therapeutic strategies and future directions are discussed, emphasizing the integration of genomic profiling and targeted interventions in managing bone metastases. The evolving landscape of genetic research, including the application of next-generation sequencing and CRISPR technology, offers promising avenues for novel and more effective therapeutic strategies. This comprehensive exploration aims to provide insights into the molecular intricacies of drug resistance in bone metastases, paving the way for improved clinical management and patient care.

骨转移是各种癌症中常见且严重的并发症,显著影响预后和生活质量。这篇综述文章深入探讨了骨转移耐药的遗传和表观遗传机制,这是有效治疗癌症的关键挑战。癌症耐药的发展可以表现为内在的或获得性的,遗传异质性起着关键作用。内在耐药通常是由于先前存在的突变,而获得性耐药是通过治疗期间的遗传和表观遗传改变而演变的。这些改变包括驱动基因如TP53和RB1的突变,表观遗传修饰如DNA甲基化和组蛋白改变,以及通路改变,特别是涉及RANK-RANKL信号和PI3K/AKT/mTOR级联。最近的研究强调了肿瘤微环境在促进耐药中的重要性,其中癌症相关成纤维细胞和缺氧等成分起着至关重要的作用。转移癌细胞与骨微环境之间的相互作用促进了耐药克隆的生存和增殖。这篇综述强调了了解这些复杂的相互作用对于开发能够克服耐药性和改善治疗结果的靶向治疗的必要性。目前的治疗策略和未来的方向进行了讨论,强调整合基因组分析和有针对性的干预管理骨转移。基因研究的不断发展,包括下一代测序和CRISPR技术的应用,为新的更有效的治疗策略提供了有希望的途径。这项全面的探索旨在为骨转移耐药的分子复杂性提供见解,为改善临床管理和患者护理铺平道路。
{"title":"The genetic architecture of bone metastases: unveiling the role of epigenetic and genetic modifications in drug resistance.","authors":"Ahmad Dawalibi, Mohamad Bakir, Khalid S Mohammad","doi":"10.20517/cdr.2025.28","DOIUrl":"https://doi.org/10.20517/cdr.2025.28","url":null,"abstract":"<p><p>Bone metastases represent frequent and severe complications in various cancers, notably impacting prognosis and quality of life. This review article delves into the genetic and epigenetic mechanisms underpinning drug resistance in bone metastases, a key challenge in effective cancer treatment. The development of drug resistance in cancer can manifest as either intrinsic or acquired, with genetic heterogeneity playing a pivotal role. Intrinsic resistance is often due to pre-existing mutations, while acquired resistance evolves through genetic and epigenetic alterations during treatment. These alterations include mutations in driver genes like <i>TP53</i> and <i>RB1</i>, epigenetic modifications such as DNA methylation and histone changes, and pathway alterations, notably involving RANK-RANKL signaling and the PI3K/AKT/mTOR cascade. Recent studies underline the significance of the tumor microenvironment in fostering drug resistance, with components such as cancer-associated fibroblasts and hypoxia playing crucial roles. The interactions between metastatic cancer cells and the bone microenvironment facilitate survival and the proliferation of drug-resistant clones. This review highlights the necessity of understanding these complex interactions to develop targeted therapies that can overcome resistance and improve treatment outcomes. Current therapeutic strategies and future directions are discussed, emphasizing the integration of genomic profiling and targeted interventions in managing bone metastases. The evolving landscape of genetic research, including the application of next-generation sequencing and CRISPR technology, offers promising avenues for novel and more effective therapeutic strategies. This comprehensive exploration aims to provide insights into the molecular intricacies of drug resistance in bone metastases, paving the way for improved clinical management and patient care.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"19"},"PeriodicalIF":4.6,"publicationDate":"2025-04-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12059479/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144029874","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
Mechanisms of resistance to NAMPT inhibitors in cancer. 癌症对NAMPT抑制剂的耐药机制。
IF 4.6 Q1 ONCOLOGY Pub Date : 2025-04-16 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2024.216
Jansen Redler, Ariana E Nelson, Christine M Heske

A common barrier to the development of effective anticancer agents is the development of drug resistance. This obstacle remains a challenge to successful clinical translation, particularly for targeted agents. Nicotinamide phosphoribosyltransferase (NAMPT) inhibitors represent a clinically applicable drug class that exploits the increased dependence of cancer cells on nicotinamide adenine dinucleotide (NAD+), a coenzyme essential to metabolism and other cellular functions. NAMPT catalyzes the rate-limiting step in the NAD+ salvage pathway of mammalian cells and is overexpressed in numerous types of cancers. Preclinical research has demonstrated that pharmacological targeting of NAMPT may be an effective strategy against certain cancers, and while several early-phase clinical trials testing NAMPT inhibitors in refractory cancers have been completed, drug resistance is a concern. Preclinical work in a variety of cancer models has demonstrated the emergence of resistance to multiple NAMPT inhibitors through several recurrent mechanisms. This review represents the first article summarizing the current state of knowledge regarding the mechanisms of acquired drug resistance to NAMPT inhibitors with a particular focus on upregulation of the compensatory NAD+ production enzymes nicotinate phosphoribosyltransferase (NAPRT) and quinolinate phosphoribosyltransferase (QPRT), acquired mutations in NAMPT, metabolic reprogramming, and altered expression of the ATP-binding cassette (ABC) efflux transporter ABCB1. An understanding of how these mechanisms interact with the biology of each given cancer cell type to predispose to the acquisition of NAMPT inhibitor resistance will be necessary to develop strategies to optimize the use of these agents moving forward.

开发有效抗癌药物的一个共同障碍是耐药性的发展。这一障碍仍然是成功临床翻译的挑战,特别是针对靶向药物。烟酰胺磷酸核糖基转移酶(NAMPT)抑制剂是一种临床适用的药物,利用癌细胞对烟酰胺腺嘌呤二核苷酸(NAD+)的依赖性增加,这是一种代谢和其他细胞功能所必需的辅酶。NAMPT在哺乳动物细胞的NAD+挽救途径中催化限速步骤,并在许多类型的癌症中过表达。临床前研究表明,NAMPT的药理学靶向可能是对抗某些癌症的有效策略,虽然一些早期临床试验已经完成,测试NAMPT抑制剂治疗难治性癌症,但耐药性是一个问题。在多种癌症模型中的临床前研究表明,通过几种复发机制出现了对多种NAMPT抑制剂的耐药性。这篇综述是第一篇总结NAMPT抑制剂获得性耐药机制的文章,特别关注代偿性NAD+产生酶烟酸磷酸核糖基转移酶(NAPRT)和喹啉酸磷酸核糖基转移酶(QPRT)的上调、NAMPT的获得性突变、代谢重编程和atp结合盒(ABC)外排转运蛋白ABCB1的表达改变。了解这些机制如何与每种给定癌细胞类型的生物学相互作用以倾向于获得NAMPT抑制剂抗性,对于制定策略以优化这些药物的使用将是必要的。
{"title":"Mechanisms of resistance to NAMPT inhibitors in cancer.","authors":"Jansen Redler, Ariana E Nelson, Christine M Heske","doi":"10.20517/cdr.2024.216","DOIUrl":"https://doi.org/10.20517/cdr.2024.216","url":null,"abstract":"<p><p>A common barrier to the development of effective anticancer agents is the development of drug resistance. This obstacle remains a challenge to successful clinical translation, particularly for targeted agents. Nicotinamide phosphoribosyltransferase (NAMPT) inhibitors represent a clinically applicable drug class that exploits the increased dependence of cancer cells on nicotinamide adenine dinucleotide (NAD<sup>+</sup>), a coenzyme essential to metabolism and other cellular functions. NAMPT catalyzes the rate-limiting step in the NAD<sup>+</sup> salvage pathway of mammalian cells and is overexpressed in numerous types of cancers. Preclinical research has demonstrated that pharmacological targeting of NAMPT may be an effective strategy against certain cancers, and while several early-phase clinical trials testing NAMPT inhibitors in refractory cancers have been completed, drug resistance is a concern. Preclinical work in a variety of cancer models has demonstrated the emergence of resistance to multiple NAMPT inhibitors through several recurrent mechanisms. This review represents the first article summarizing the current state of knowledge regarding the mechanisms of acquired drug resistance to NAMPT inhibitors with a particular focus on upregulation of the compensatory NAD<sup>+</sup> production enzymes nicotinate phosphoribosyltransferase (NAPRT) and quinolinate phosphoribosyltransferase (QPRT), acquired mutations in NAMPT, metabolic reprogramming, and altered expression of the ATP-binding cassette (ABC) efflux transporter ABCB1. An understanding of how these mechanisms interact with the biology of each given cancer cell type to predispose to the acquisition of NAMPT inhibitor resistance will be necessary to develop strategies to optimize the use of these agents moving forward.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"18"},"PeriodicalIF":4.6,"publicationDate":"2025-04-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12059476/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144055081","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
Exosomal miR-92b-5p regulates N4BP1 to enhance PTEN mono-ubiquitination in doxorubicin-resistant AML. 外泌体miR-92b-5p调节N4BP1增强抗阿霉素AML中PTEN单泛素化。
IF 4.6 Q1 ONCOLOGY Pub Date : 2025-03-28 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2024.140
Qianyuan Li, Jie Cheng, Danni Qin, Sheng Xiao, Chenjiao Yao

Aim: Doxorubicin, pivotal for acute myeloid leukemia (AML) treatment, often succumbs to resistance, impeding therapeutic success. Although exosomal transfer is linked to chemoresistance, the detailed role of exosomal miRNAs in doxorubicin resistance remains incompletely understood. Methods: We employed miRNA sequencing to delineate the profile of exosomal miRNAs in doxorubicin-resistant K562/DOX cells and AML patients. Subsequently, qPCR was utilized to scrutinize the expression of exosomal miR-92b-5p in these resistant cells and AML patients. A dual-luciferase reporter assay was conducted to elucidate the direct binding of miR-92b-5p to NEDD4 binding protein 1 (N4BP1). Furthermore, interactions between N4BP1 and NEDD4, as well as between NEDD4 and PTEN, were investigated by co-immunoprecipitation (Co-IP). Meanwhile, the ubiquitination of PTEN was also examined by Co-IP. Western blot analysis was applied to assess the expression levels of N4BP1, NEDD4, PTEN, RAD51, and proteins associated with the PI3K-AKT-mTOR pathway. Gain- and loss-of-function studies were conducted to ascertain the functional role of miR-92b-5p in doxorubicin resistance by using miR-92b-5p-mimic and miR-92b-5p-inhibitor transfections. Results: Our study found exosomal miR-92b-5p was upregulated both in doxorubicin-resistant cells and AML patients. Moreover, miR-92b-5p targets N4BP1, promoting NEDD4-mediated mono-ubiquitination of PTEN. This alters PTEN's subcellular localization, promoting nuclear PTEN and reducing cytoplasmic PTEN, which in turn leads to increased RAD51 for DNA repair and activation of the PI3K-AKT-mTOR pathway for cell proliferation, contributing to doxorubicin resistance. Conclusion: Our study reveals a novel mechanism of doxorubicin resistance mediated by exosomal miR-92b-5p and provides potential therapeutic targets for overcoming drug resistance in AML.

目的:阿霉素是治疗急性髓性白血病(AML)的关键药物,但它经常出现耐药性,阻碍了治疗的成功。尽管外泌体转移与化疗耐药有关,但外泌体mirna在阿霉素耐药中的详细作用仍不完全清楚。方法:我们采用miRNA测序来描述阿霉素耐药K562/DOX细胞和AML患者的外泌体miRNA谱。随后,利用qPCR检查这些耐药细胞和AML患者中外泌体miR-92b-5p的表达。通过双荧光素酶报告基因实验,我们阐明了miR-92b-5p与NEDD4结合蛋白1 (N4BP1)的直接结合。此外,通过共免疫沉淀(Co-IP)研究了N4BP1与NEDD4以及NEDD4与PTEN之间的相互作用。同时,通过Co-IP检测PTEN的泛素化。Western blot检测N4BP1、NEDD4、PTEN、RAD51以及PI3K-AKT-mTOR通路相关蛋白的表达水平。通过转染miR-92b-5p mimic和miR-92b-5p inhibitor,进行了功能获得和功能丧失研究,以确定miR-92b-5p在阿霉素耐药中的功能作用。结果:我们的研究发现,在阿霉素耐药细胞和AML患者中,外泌体miR-92b-5p均上调。此外,miR-92b-5p靶向N4BP1,促进nedd4介导的PTEN单泛素化。这改变了PTEN的亚细胞定位,促进核PTEN和减少细胞质PTEN,进而导致用于DNA修复的RAD51增加和用于细胞增殖的PI3K-AKT-mTOR通路的激活,从而导致阿霉素耐药。结论:我们的研究揭示了外泌体miR-92b-5p介导的阿霉素耐药的新机制,并为克服AML耐药提供了潜在的治疗靶点。
{"title":"Exosomal miR-92b-5p regulates N4BP1 to enhance PTEN mono-ubiquitination in doxorubicin-resistant AML.","authors":"Qianyuan Li, Jie Cheng, Danni Qin, Sheng Xiao, Chenjiao Yao","doi":"10.20517/cdr.2024.140","DOIUrl":"10.20517/cdr.2024.140","url":null,"abstract":"<p><p><b>Aim:</b> Doxorubicin, pivotal for acute myeloid leukemia (AML) treatment, often succumbs to resistance, impeding therapeutic success. Although exosomal transfer is linked to chemoresistance, the detailed role of exosomal miRNAs in doxorubicin resistance remains incompletely understood. <b>Methods:</b> We employed miRNA sequencing to delineate the profile of exosomal miRNAs in doxorubicin-resistant K562/DOX cells and AML patients. Subsequently, qPCR was utilized to scrutinize the expression of exosomal miR-92b-5p in these resistant cells and AML patients. A dual-luciferase reporter assay was conducted to elucidate the direct binding of miR-92b-5p to NEDD4 binding protein 1 (N4BP1). Furthermore, interactions between N4BP1 and NEDD4, as well as between NEDD4 and PTEN, were investigated by co-immunoprecipitation (Co-IP). Meanwhile, the ubiquitination of PTEN was also examined by Co-IP. Western blot analysis was applied to assess the expression levels of N4BP1, NEDD4, PTEN, RAD51, and proteins associated with the PI3K-AKT-mTOR pathway. Gain- and loss-of-function studies were conducted to ascertain the functional role of miR-92b-5p in doxorubicin resistance by using miR-92b-5p-mimic and miR-92b-5p-inhibitor transfections. <b>Results:</b> Our study found exosomal miR-92b-5p was upregulated both in doxorubicin-resistant cells and AML patients. Moreover, miR-92b-5p targets N4BP1, promoting NEDD4-mediated mono-ubiquitination of PTEN. This alters PTEN's subcellular localization, promoting nuclear PTEN and reducing cytoplasmic PTEN, which in turn leads to increased RAD51 for DNA repair and activation of the PI3K-AKT-mTOR pathway for cell proliferation, contributing to doxorubicin resistance. <b>Conclusion:</b> Our study reveals a novel mechanism of doxorubicin resistance mediated by exosomal miR-92b-5p and provides potential therapeutic targets for overcoming drug resistance in AML.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"16"},"PeriodicalIF":4.6,"publicationDate":"2025-03-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11977356/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143813085","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
Circular RNAs modulate cancer drug resistance: advances and challenges. 环状rna调节癌症耐药:进展和挑战。
IF 4.6 Q1 ONCOLOGY Pub Date : 2025-03-28 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2024.195
Jinghan Hua, Zhe Wang, Xiaoxun Cheng, Jiaojiao Dai, Ping Zhao

Acquired drug resistance is a main factor contributing to cancer therapy failure and high cancer mortality, highlighting the necessity to develop novel intervention targets. Circular RNAs (circRNAs), an abundant class of RNA molecules with a closed loop structure, possess characteristics including high stability, which provide unique advantages in clinical application. Growing evidence indicates that aberrantly expressed circRNAs are associated with resistance against various cancer treatments, including targeted therapy, chemotherapy, radiotherapy, and immunotherapy. Therefore, targeting these aberrant circRNAs may offer a strategy to improve the efficiency of cancer therapy. Herein, we present a summary of the most recently studied circRNAs and their regulatory roles on cancer drug resistance. With the advances in artificial intelligence (AI)-based bioinformatics algorithms, circRNAs could emerge as promising biomarkers and intervention targets in cancer therapy.

获得性耐药性是导致癌症治疗失败和癌症死亡率居高不下的一个主要因素,这凸显了开发新型干预靶点的必要性。环状 RNA(circRNA)是一类丰富的具有闭环结构的 RNA 分子,具有高稳定性等特点,在临床应用中具有独特的优势。越来越多的证据表明,异常表达的 circRNA 与各种癌症治疗(包括靶向治疗、化疗、放疗和免疫疗法)的耐药性有关。因此,针对这些异常的 circRNAs 可能是提高癌症治疗效率的一种策略。在此,我们总结了最新研究的circRNAs及其对癌症耐药性的调控作用。随着以人工智能(AI)为基础的生物信息学算法的进步,circRNAs可能会成为癌症治疗中前景广阔的生物标记物和干预靶点。
{"title":"Circular RNAs modulate cancer drug resistance: advances and challenges.","authors":"Jinghan Hua, Zhe Wang, Xiaoxun Cheng, Jiaojiao Dai, Ping Zhao","doi":"10.20517/cdr.2024.195","DOIUrl":"10.20517/cdr.2024.195","url":null,"abstract":"<p><p>Acquired drug resistance is a main factor contributing to cancer therapy failure and high cancer mortality, highlighting the necessity to develop novel intervention targets. Circular RNAs (circRNAs), an abundant class of RNA molecules with a closed loop structure, possess characteristics including high stability, which provide unique advantages in clinical application. Growing evidence indicates that aberrantly expressed circRNAs are associated with resistance against various cancer treatments, including targeted therapy, chemotherapy, radiotherapy, and immunotherapy. Therefore, targeting these aberrant circRNAs may offer a strategy to improve the efficiency of cancer therapy. Herein, we present a summary of the most recently studied circRNAs and their regulatory roles on cancer drug resistance. With the advances in artificial intelligence (AI)-based bioinformatics algorithms, circRNAs could emerge as promising biomarkers and intervention targets in cancer therapy.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"17"},"PeriodicalIF":4.6,"publicationDate":"2025-03-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11977347/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143813068","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
Strategies to combat cancer drug resistance: focus on copper metabolism and cuproptosis. 对抗癌症耐药的策略:关注铜代谢和铜还原。
IF 4.6 Q1 ONCOLOGY Pub Date : 2025-03-26 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.41
Leyi Yao, Baoyi Jiang, Dacai Xu

Cancer cells often develop tolerance to chemotherapy, targeted therapy, and immunotherapy drugs either before or during treatment. The significant heterogeneity among various tumors poses a critical challenge in modern cancer research, particularly in overcoming drug resistance. Copper, as an essential trace element in the body, participates in various biological processes of diseases, including cancers. The growth of many types of tumor cells exhibits a heightened dependence on copper. Thus, targeting copper metabolism or inducing cuproptosis may be potential ways to overcome cancer drug resistance. Copper chelators have shown potential in overcoming cancer drug resistance by targeting copper-dependent processes in cancer cells. In contrast, copper ionophores, copper-based nanomaterials, and other small molecules have been used to induce copper-dependent cell death (cuproptosis) in cancer cells, including drug-resistant tumor cells. This review summarizes the regulation of copper metabolism and cuproptosis in cancer cells and the role of copper metabolism and cuproptosis in cancer drug resistance, providing ideas for overcoming cancer resistance in the future.

癌细胞通常在治疗前或治疗过程中对化疗、靶向治疗和免疫治疗药物产生耐受性。各种肿瘤之间的显著异质性对现代癌症研究,特别是克服耐药性提出了重大挑战。铜作为人体必需的微量元素,参与包括癌症在内的各种疾病的生物过程。许多类型的肿瘤细胞的生长表现出对铜的高度依赖。因此,靶向铜代谢或诱导铜还原可能是克服癌症耐药的潜在途径。铜螯合剂通过靶向癌细胞中的铜依赖过程,显示出克服癌症耐药的潜力。相比之下,铜离子载体、铜基纳米材料和其他小分子已被用于诱导癌症细胞(包括耐药肿瘤细胞)中的铜依赖性细胞死亡(铜增生)。本文综述了肿瘤细胞中铜代谢和铜沉淀的调控以及铜代谢和铜沉淀在肿瘤耐药中的作用,为今后克服肿瘤耐药提供思路。
{"title":"Strategies to combat cancer drug resistance: focus on copper metabolism and cuproptosis.","authors":"Leyi Yao, Baoyi Jiang, Dacai Xu","doi":"10.20517/cdr.2025.41","DOIUrl":"10.20517/cdr.2025.41","url":null,"abstract":"<p><p>Cancer cells often develop tolerance to chemotherapy, targeted therapy, and immunotherapy drugs either before or during treatment. The significant heterogeneity among various tumors poses a critical challenge in modern cancer research, particularly in overcoming drug resistance. Copper, as an essential trace element in the body, participates in various biological processes of diseases, including cancers. The growth of many types of tumor cells exhibits a heightened dependence on copper. Thus, targeting copper metabolism or inducing cuproptosis may be potential ways to overcome cancer drug resistance. Copper chelators have shown potential in overcoming cancer drug resistance by targeting copper-dependent processes in cancer cells. In contrast, copper ionophores, copper-based nanomaterials, and other small molecules have been used to induce copper-dependent cell death (cuproptosis) in cancer cells, including drug-resistant tumor cells. This review summarizes the regulation of copper metabolism and cuproptosis in cancer cells and the role of copper metabolism and cuproptosis in cancer drug resistance, providing ideas for overcoming cancer resistance in the future.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"15"},"PeriodicalIF":4.6,"publicationDate":"2025-03-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11977383/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143813093","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
期刊
癌症耐药(英文)
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
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
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1