癌症耐药(英文)最新文献

筛选
英文 中文
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
癌症耐药(英文) 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
{"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
癌症耐药(英文) Pub Date : 2025-04-24 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.14
Jinglu Yu, Xiaoni Kong, Yu Feng
{"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
癌症耐药(英文) 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
{"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
癌症耐药(英文) Pub Date : 2025-04-22 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.28
Ahmad Dawalibi, Mohamad Bakir, Khalid S Mohammad
{"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
癌症耐药(英文) Pub Date : 2025-04-16 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2024.216
Jansen Redler, Ariana E Nelson, Christine M Heske
{"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
癌症耐药(英文) 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
{"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
癌症耐药(英文) 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
{"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
癌症耐药(英文) Pub Date : 2025-03-26 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2025.41
Leyi Yao, Baoyi Jiang, Dacai Xu
{"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
Unraveling BRAF alterations: molecular insights to circumvent therapeutic resistance across cancer types. 揭示BRAF改变:规避癌症类型治疗耐药性的分子洞察。
IF 4.6
癌症耐药(英文) Pub Date : 2025-03-24 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2024.213
Caterina Perfetto, Marianna Aprile, Simona Cataldi, Elisa Giovannetti, Valerio Costa
{"title":"Unraveling <i>BRAF</i> alterations: molecular insights to circumvent therapeutic resistance across cancer types.","authors":"Caterina Perfetto, Marianna Aprile, Simona Cataldi, Elisa Giovannetti, Valerio Costa","doi":"10.20517/cdr.2024.213","DOIUrl":"10.20517/cdr.2024.213","url":null,"abstract":"<p><p><b>Aim:</b> As intrinsic resistance - often driven by concurrent genomic alterations in tumor suppressor genes or oncogenes - remains a major challenge in oncology, this work aimed to comprehensively analyze <i>BRAF</i> somatic alterations across cancer types and identify new potential therapeutic strategies to overcome drug resistance. <b>Methods:</b> We conducted an extensive analysis of genomics, transcriptomics, and clinical data retrieved from public repositories, including cBioPortal. Our comprehensive analysis examined <i>BRAF</i> alterations [point mutations, structural variants (SVs) and copy number alteration] in more than 217,000 tumor samples across 120 distinct tumor types from primary and metastatic sites in both adult and pediatric cohorts, focusing on mutual exclusivity and co-occurrence of mutations in other oncogenes or tumor suppressors. The work also explores the association of <i>BRAF</i> somatic alterations with survival, clinical and molecular features. <b>Results:</b> Analysis of mutation frequencies across cancer types revealed that BRAFV600E represents approximately 90% of all <i>BRAF</i> alterations. While melanoma and thyroid carcinoma show the highest prevalence of <i>BRAF</i> mutations, followed by colorectal and non-small cell lung cancer in terms of absolute number of patients harboring <i>BRAF</i> mutations worldwide, notably high mutation frequencies were identified in rare malignancies, including hairy-cell leukemia, ganglioglioma, and serous borderline ovarian tumors. The comprehensive analysis of genomic profiling data across these tumors uncovered distinct patterns of co-occurring and mutually exclusive alterations in oncogenes and tumor suppressor genes, illuminating resistance mechanisms and suggesting novel therapeutic combinations. <b>Conclusion:</b> Comprehensive genomic profiling is critical for optimizing targeted therapy and overcoming drug resistance in <i>BRAF</i>-mutated cancers. The identification of co-occurring alterations provides opportunities for rational combination therapies, emphasizing the importance of detailed mutation profiling in developing effective treatment strategies across diverse cancer types.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"14"},"PeriodicalIF":4.6,"publicationDate":"2025-03-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11977354/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143813123","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
Research advances in natural sesquiterpene lactones: overcoming cancer drug resistance through modulation of key signaling pathways. 天然倍半萜内酯的研究进展:通过调节关键信号通路克服癌症耐药。
IF 4.6
癌症耐药(英文) Pub Date : 2025-03-24 eCollection Date: 2025-01-01 DOI: 10.20517/cdr.2024.178
Chi Teng, Jia-Wen Chen, Li-Sha Shen, Sibao Chen, Guo-Qing Chen
{"title":"Research advances in natural sesquiterpene lactones: overcoming cancer drug resistance through modulation of key signaling pathways.","authors":"Chi Teng, Jia-Wen Chen, Li-Sha Shen, Sibao Chen, Guo-Qing Chen","doi":"10.20517/cdr.2024.178","DOIUrl":"10.20517/cdr.2024.178","url":null,"abstract":"<p><p>Cancer remains a significant global health challenge, with current chemotherapeutic strategies frequently limited by the emergence of resistance. In this context, natural compounds with the potential to overcome resistance have garnered considerable attention. Among these, sesquiterpene lactones, primarily derived from plants in the Asteraceae family, stand out for their potential anticancer properties. This review specifically focuses on five key signaling pathways: PI3K/Akt/mTOR, NF-κB, Wnt/β-catenin, MAPK/ERK, and STAT3, which play central roles in the mechanisms of cancer resistance. For each of these pathways, we detail their involvement in both cancer development and the emergence of drug resistance. Additionally, we investigate how sesquiterpene lactones modulate these pathways to overcome resistance across diverse cancer types. These insights highlight the potential of sesquiterpene lactones to drive the advancement of novel therapies that can effectively combat both cancer progression and drug resistance.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"8 ","pages":"13"},"PeriodicalIF":4.6,"publicationDate":"2025-03-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11977367/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143813088","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:481959085
Book学术官方微信