Cancer Biology & TherapyPub Date : 2025-12-01Epub Date: 2025-03-13DOI: 10.1080/15384047.2025.2475581
Sana Noreen, Izwa Ishaq, Muhammad Hamzah Saleem, Baber Ali, Syed Muhammad Ali, Javed Iqbal
{"title":"Electrochemical biosensing in oncology: a review advancements and prospects for cancer diagnosis.","authors":"Sana Noreen, Izwa Ishaq, Muhammad Hamzah Saleem, Baber Ali, Syed Muhammad Ali, Javed Iqbal","doi":"10.1080/15384047.2025.2475581","DOIUrl":"10.1080/15384047.2025.2475581","url":null,"abstract":"<p><p>Early and precise diagnosis of cancer is pivotal for effective therapeutic intervention. Traditional diagnostic methods, despite their reliability, often face limitations such as invasiveness, high costs, labor-intensive procedures, extended processing times, and reduced sensitivity for early-stage detection. Electrochemical biosensing is a revolutionary method that provides rapid, cost-effective, and highly sensitive detection of cancer biomarkers. This review discusses the use of electrochemical detection in biosensors to provide real-time insights into disease-specific molecular interactions, focusing on target recognition and signal generation mechanisms. Furthermore, the superior efficacy of electrochemical biosensors compared to conventional techniques is explored, particularly in their ability to detect cancer biomarkers with enhanced specificity and sensitivity. Advancements in electrode materials and nanostructured designs, integrating nanotechnology, microfluidics, and artificial intelligence, have the potential to overcome biological interferences and scale for clinical use. Research and innovation in oncology diagnostics hold potential for personalized medicine, despite challenges in commercial viability and real-world application.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"26 1","pages":"2475581"},"PeriodicalIF":4.4,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11913392/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143613291","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer Biology & TherapyPub Date : 2025-12-01Epub Date: 2025-01-29DOI: 10.1080/15384047.2025.2459426
Zhanglin Zhang, Xiang Lin, Yaling Yang, Xuemei Wang, Yi Wang, Xianbao Huang, Miao Hong, Wei Gao, Hua He, M James You, Yi Yang, Guangyao Kong
{"title":"Caspase 3-specific cleavage of ubiquitin-specific peptidase 48 enhances drug-induced apoptosis in AML.","authors":"Zhanglin Zhang, Xiang Lin, Yaling Yang, Xuemei Wang, Yi Wang, Xianbao Huang, Miao Hong, Wei Gao, Hua He, M James You, Yi Yang, Guangyao Kong","doi":"10.1080/15384047.2025.2459426","DOIUrl":"10.1080/15384047.2025.2459426","url":null,"abstract":"<p><p>Dysfunction or dysregulation of deubiquitination is closely related to the initiation and development of multiple cancers. Targeted regulation of deubiquitination has been recognized as an important strategy in tumor therapy. However, the mechanism by which drugs regulate deubiquitinase is not clear. Here, we identified ubiquitin-specific peptidase 48 (USP48), a member of the ubiquitin-specific protease family highly expressed in various tumors, as a specific substrate for the activated caspase-3. During drug induced apoptosis of AML cells, activated caspase-3 cleaves USP48 through recognizing the conservative motif DEQD located at 611-614 sites of human USP48. Subsequent analysis showed that the cleavage USP48 N-terminal fragment which contains catalytic active domain is easily degraded by ubiquitination. Meanwhile knockdown experiment showed that inhibiting the expression of USP48 could also promotes apoptosis and enhance the efficacy of chemotherapy drugs. Altogether, these results suggest that targeting USP48 may represent a novel therapeutic strategy in AML.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"26 1","pages":"2459426"},"PeriodicalIF":4.4,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11781246/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143058280","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer Biology & TherapyPub Date : 2025-12-01Epub Date: 2025-08-13DOI: 10.1080/15384047.2025.2541974
Larissa Akemi Kido, Milena Rodrigues Marusco, Ellen Aparecida da Silva, Laís Do Carmo, Ana Beatriz Teodoro Borges, Felipe Luz Torres Silva, Juliana Silveira Ruas, Dieila Giomo de Lima, Larissa de Abreu Fernandes, Camila Maia Martin Daiggi, Izilda Aparecida Cardinalli, Mayara Ferreira Euzébio, Patricia Yoshioka Jotta, Mariana Maschietto, Priscila Pini Zenatti
{"title":"Establishing a pediatric solid tumor PDX biobank for precision oncology research.","authors":"Larissa Akemi Kido, Milena Rodrigues Marusco, Ellen Aparecida da Silva, Laís Do Carmo, Ana Beatriz Teodoro Borges, Felipe Luz Torres Silva, Juliana Silveira Ruas, Dieila Giomo de Lima, Larissa de Abreu Fernandes, Camila Maia Martin Daiggi, Izilda Aparecida Cardinalli, Mayara Ferreira Euzébio, Patricia Yoshioka Jotta, Mariana Maschietto, Priscila Pini Zenatti","doi":"10.1080/15384047.2025.2541974","DOIUrl":"10.1080/15384047.2025.2541974","url":null,"abstract":"<p><p>Developing advanced preclinical models and targeted therapies is essential for reducing cancer-related deaths in children with solid tumors. Patient-derived xenografts (PDX) have the potential to replicate key elements of the original tumor, including morphology, genetic alterations, and microenvironment, making them valuable tools for studying tumor biology and drug response. We implanted 124 pediatric solid tumor samples, collected for 1 y, into NOD/SCID/IL2Rg (NSG) mice. Tumor fragments were placed subcutaneously, and the animals were monitored for up to 1 y. Histopathology, Short Tandem Repeat (STR) profiling, RT-PCR and/or RNA-sequencing were performed to confirm tumor identity and detect driver fusions. Fifty-five xenografts were successfully established (44.35% of implanted samples), representing 19 tumor types. Sarcomas, notably osteosarcoma, Ewing sarcoma, synovial sarcoma, and rhabdomyosarcoma, displayed first-generation engraftment rates above 55%. Central nervous system tumors had lower success, reflecting unique microenvironmental requirements. Histopathology and STR concordances were 85.45% and 81.1%, respectively, while 92.6% of sarcoma PDXs retained original fusion genes. Second-generation xenografts showed faster growth, suggesting adaptation to the murine host. Sporadic discrepancies, such as new fusions or lymphoproliferative expansions, indicated the need for ongoing molecular validation parallel to other techniques. A pediatric PDX biobank can effectively capture key tumor features while facilitating the study of therapeutic responses and tumor evolution. Our models confirm the feasibility of achieving stable histological and molecular profiles, offering a valuable resource for precision oncology research. Ultimately, these pediatric PDXs could accelerate the discovery of targeted therapy and significantly improve treatment outcomes.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"26 1","pages":"2541974"},"PeriodicalIF":4.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12351738/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144834058","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer Biology & TherapyPub Date : 2025-12-01Epub Date: 2025-06-21DOI: 10.1080/15384047.2025.2510029
Chao-Bang Xie, Zi-Jian Tang, Yang Wu, Kai-Fei Zhao
{"title":"Unresectable ectopic hepatocellular carcinoma in the left sub-diaphragmatic area and retroperitoneum treated with TACE: case report and literature review.","authors":"Chao-Bang Xie, Zi-Jian Tang, Yang Wu, Kai-Fei Zhao","doi":"10.1080/15384047.2025.2510029","DOIUrl":"10.1080/15384047.2025.2510029","url":null,"abstract":"<p><p>Ectopic hepatocellular carcinoma (EHCC) demonstrates morphological and immunohistochemical features identical to conventional intrahepatic hepatocellular carcinoma (HCC), originating from ectopic liver tissue (EL), which is defined as a hepatic organ or tissue not connected to the mother liver. EHCC is a rare disease and difficult to diagnose preoperatively. Therefore, its epidemiology, treatment, and prognosis are not fully elucidated. Herein, we present a case report of a 52-year-old male diagnosed with unresectable EHCC who underwent transarterial chemoembolization (TACE) and succumbed to the disease 13 months after treatment, accompanied by a literature review summarizing the epidemiology while analyzing therapeutic strategies and prognostic outcomes in EHCC through synthesis of published case evidence.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"26 1","pages":"2510029"},"PeriodicalIF":4.4,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144339915","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer Biology & TherapyPub Date : 2025-12-01Epub Date: 2025-08-13DOI: 10.1080/15384047.2025.2546219
Zhuo Li, Qinghua Jiang, Quan Yang, Yujie Zhou, Jiansong Wang
{"title":"Hypoxia-induced PYCR1 regulates glycolysis and histone lactylation to promote bladder cancer progression and metastasis via SLC6A14/Glutamine metabolism.","authors":"Zhuo Li, Qinghua Jiang, Quan Yang, Yujie Zhou, Jiansong Wang","doi":"10.1080/15384047.2025.2546219","DOIUrl":"10.1080/15384047.2025.2546219","url":null,"abstract":"<p><p>Hypoxia-induced Pyrroline-5-Carboxylate Reductase 1 (PYCR1) is implicated in bladder cancer (BC), but its specific role remains elusive. This study investigated how PYCR1 promotes BC progression through glycolysis, histone H3 Lysine 18 Lactylation (H3K18la), and Solute Carrier Family 6 Member 14 (SLC6A14)-driven glutamine catabolism. Here, BC cell lines were cultured under hypoxia to evaluate changes in PYCR1 expression, glycolysis, and lactate production. The xenograft and metastasis models in nude mice were used to validate the role of the PYCR1/H3K18la/SLC6A14 axis in BC progression. GEPIA Bioinformatics database data showed that PYCR1 was upregulated in BC and was associated with poor prognosis. The PYCR1 positive expression rate in BC tissues was increased. Hypoxia induced PYCR1 expression in BC cells, enhancing glycolysis and lactate production, which increased H3K18la levels. Upregulated SLC6A14 expression promoted glutamine catabolism and enhanced BC cell proliferation, migration, and invasion. PYCR1 knockdown inhibited H3K18la levels, SLC6A14 expression, and BC cell aggressiveness; SLC6A14 overexpression reversed these effects. <i>In vivo</i> experiments confirmed that the PYCR1/H3K18la/SLC6A14 axis is critical for hypoxia-driven BC growth and metastasis. In summary, Hypoxia-induced PYCR1 enhances glycolysis, leading to increased lactate production and elevated H3K18la levels, which upregulates SLC6A14 transcription and glutamine catabolism, thereby promoting BC growth and metastasis.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"26 1","pages":"2546219"},"PeriodicalIF":4.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12355718/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144844468","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer Biology & TherapyPub Date : 2025-12-01Epub Date: 2025-04-23DOI: 10.1080/15384047.2025.2495999
Yayun Cui, Xiaofeng Zhu, Liting Qian, Shu Zhang
{"title":"Extracellular transfer of HuR promotes acquired cisplatin resistance in esophageal cancer cells.","authors":"Yayun Cui, Xiaofeng Zhu, Liting Qian, Shu Zhang","doi":"10.1080/15384047.2025.2495999","DOIUrl":"https://doi.org/10.1080/15384047.2025.2495999","url":null,"abstract":"<p><p>Cisplatin (DDP) resistance is a key factor hindering esophageal cancer (ESCA) treatment. Exosomes have been reported to confer resistance to DDP in various tumor cells. However, the effects of ESCA cell-derived exosomes and exosomal human antigen R (HuR) on DDP resistance in cancer cells have not been elucidated. In this study, isolated exosomes were identified by transmission electron microscopy, nanoparticle tracking analysis, and western blotting. CCK-8 and flow cytometry were employed to assess the functional role of exosomes in ESCA DDP-resistant cells and their parental cells. Bioinformatics analysis was performed to identify molecules that were positively associated with HuR and validated using dual-luciferase reporter analysis and RNA immunoprecipitation assays. We found that exosomes from ESCA cells enhance the resistance of drug-resistant cells to DDP. Importantly, HuR protein, but not mRNA, was directly transferred into DDP-resistant cells via exosomes, thereby increasing the level of HuR protein. Mechanistically, HuR positively correlated with Lamin B2 (LMNB2) in ESCA cells, and ESCA DDP-resistant cells transfected with siRNA targeting LMNB2 exhibited reduced cell viability and elevated apoptosis rates. Moreover, the role of ESCA cell-derived exosomes in the transmission of DDP resistance <i>in vivo</i> was validated using a nude mouse model. Collectively, our results revealed that exosomes exposed to ESCA cells induced greater drug resistance in DDP-resistant ESCA cells via HuR delivery. Targeting HuR or its positively related target LMNB2 may present new therapeutic opportunities for treating patients with DDP-resistant ESCA.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"26 1","pages":"2495999"},"PeriodicalIF":4.4,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12026070/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143980559","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"miR-301a-mediated crosstalk between the Hedgehog and HIPPO/YAP signaling pathways promotes pancreatic cancer.","authors":"Bing Qi, Yuqiong Wang, Xian Zhu, Yanfang Gong, Jing Jin, Hongyu Wu, Xiaohua Man, Feng Liu, Wenzhu Yao, Jun Gao","doi":"10.1080/15384047.2025.2457761","DOIUrl":"10.1080/15384047.2025.2457761","url":null,"abstract":"<p><p>Pancreatic ductal adenocarcinoma (PDAC) poses a significant challenge in oncology due to its dismal prognosis and limited therapeutic options. In this study, we investigated the role of miR-301a in facilitating crosstalk between the Hedgehog (Hh) and HIPPO/YAP signaling pathways during the progression of PDAC. Our findings revealed that miR-301a served as a central regulatory node, targeting Gli1 within the Hh pathway and STK4 within the HIPPO/YAP pathway. Immunohistochemical and molecular analyses confirmed dysregulation of pathway components in pancreatic cancer, underscoring the pivotal role of miR-301a. Functional assays demonstrated the impact of miR-301a on cell proliferation and apoptosis, particularly in synergy with TNF-α. Overall, our study elucidated the intricate interplay between the Hh and HIPPO/YAP pathways mediated by miR-301a, providing valuable insights into potential therapeutic strategies for intervening in PDAC.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"26 1","pages":"2457761"},"PeriodicalIF":4.4,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11760222/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143058283","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer Biology & TherapyPub Date : 2025-12-01Epub Date: 2025-03-15DOI: 10.1080/15384047.2025.2472432
Cory Fines, Helen McCarthy, Niamh Buckley
{"title":"The search for a TNBC vaccine: the guardian vaccine.","authors":"Cory Fines, Helen McCarthy, Niamh Buckley","doi":"10.1080/15384047.2025.2472432","DOIUrl":"10.1080/15384047.2025.2472432","url":null,"abstract":"<p><p>Nearly 20 million people are diagnosed with cancer each year with breast cancer being the most common among women. Triple negative breast cancer (TNBC), defined by its no/low expression of ER and PR and lack of amplification of HER2, makes up 15-20% of all breast cancer cases. While patients overall have a higher response to chemotherapy, this subgroup is associated with the lowest survival rate indicating significant clinical and molecular heterogeneity demanding alternate treatment options. Therefore, new therapies have been explored, with a large focus on utilizing the immune system. A whole host of immunotherapies have been studied including immune checkpoint inhibitors, now standard of care for eligible patients, and possibly the most exciting and promising is that of a TNBC vaccine. While currently there are no approved TNBC vaccines, this review highlights many promising studies and points to an antigen, p53, which we believe is highly relevant for TNBC.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"26 1","pages":"2472432"},"PeriodicalIF":4.4,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11913391/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143633534","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"CircATP5C1 promotes triple-negative breast cancer progression by binding IGF2BP2 to modulate CSF-1 secretion.","authors":"Hongbo Liu, Haoqi Wang, Wei Gao, Yang Yuan, Tiantian Tang, Meixiang Sang, Fei Liu, Cuizhi Geng","doi":"10.1080/15384047.2025.2479926","DOIUrl":"10.1080/15384047.2025.2479926","url":null,"abstract":"<p><p>Triple-negative breast cancer (TNBC) is a common malignant disease among females and severely threatens the health of women worldwide. Nowadays, circular RNAs (circRNAs) aroused our interest for their functions in human cancers, including TNBC. However, the mechanism of most circRNAs in the progression of TNBC remains unclear. We found a novel circRNA named circATP5C1, whose function in TNBC remains uncovered. Tissue microarray was used to analyze the association between the expression of circATP5C1 and the prognoses of TNBC patients. Gain-and loss-of-function experiments were performed to validate the biological functions of circATP5C1 in different TNBC cell lines. RNA-seq analyses were conducted to find out the target genes regulated by circATP5C1. RNA pull-down assay and mass spectrometry were used to select the proteins associated with circATP5C1. RNA FISH-immunofluorescence and RNA immunoprecipitation (RIP) were complemented to validate the interaction between circATP5C1 and its binding protein. CircATP5C1 was identified to have predictive function in prognosis of TNBC patients. CircATP5C1 advanced the progression of TNBC cells. Mechanistically, Colony stimulating factor 1 (CSF-1) is a vital downstream gene regulated by circATP5C1. The alteration of CSF-1 expression level was validated due to the interaction between circATP5C1 and insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2). Rescue experiments demonstrated that circATP5C1 accelerates the progression of TNBC partly via binding with IGF2BP2 to increase the secretion of CSF-1. This study uncovers a novel mechanism of circATP5C1/IGF2BP2/CSF-1 pathway in regulating progression of TNBC.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"26 1","pages":"2479926"},"PeriodicalIF":4.4,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11980513/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143771416","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cancer Biology & TherapyPub Date : 2025-12-01Epub Date: 2025-02-24DOI: 10.1080/15384047.2025.2471631
Chan-Woong Jung, Jeong-Yub Kim, Myung-Jin Park
{"title":"CXCR4 confers stemness and radioresistance in chordoma cells.","authors":"Chan-Woong Jung, Jeong-Yub Kim, Myung-Jin Park","doi":"10.1080/15384047.2025.2471631","DOIUrl":"10.1080/15384047.2025.2471631","url":null,"abstract":"<p><p>CXC Chemokine receptor type 4 (CXCR4) is commonly considered a potential marker for cancer stem cells (CSCs). Dedifferentiated-type chordoma (DTC) cells derived from a patient with recurrent chordoma exhibit high CXCR4 expression and demonstrate increased resistance to chemotherapeutic drugs and ionizing radiation (IR) compared to the conventional-type chordoma cell line, U-CH1. However, the precise role of CXCR4 in the stemness and IR resistance of DTC remains unclear. Therefore, this study aims to elucidate the correlation between the expression of CXCR4 and stemness and radioresistance in chordoma. DTC cells expressing CXCR4 (CXCR4<sup>+</sup> DTC cells), isolated by magnetic-activated cell sorting, exhibited increased self-renewal activity, tumorigenicity, and IR resistance, accompanied by elevated Sox2 expression. Knockdown of CXCR4 expression using short hairpin RNA, inhibition of CXCR4 signaling with AMD3100, and targeting of STAT3, a downstream effector of CXCR4, with WP1066 in DTC cells significantly diminished their self-renewal ability, tumorigenic potential, IR resistance, and Sox2 expression. Additionally, transfection with a small interfering Sox2 RNA suppressed self-renewal activity, tumorigenicity, and IR resistance in DTC cells, whereas overexpression of CXCR4 reversed these effects in U-CH1 cells. Furthermore, DTC cells infected with shCXCR4 exhibited substantial tumor suppression, and the combination of IR and AMD3100 significantly reduced DTC tumor growth in a mouse xenograft model. These findings underscore the functional significance of CXCR4 as a CSC marker, highlighting its potential as a therapeutic target for malignant chordomas.</p>","PeriodicalId":9536,"journal":{"name":"Cancer Biology & Therapy","volume":"26 1","pages":"2471631"},"PeriodicalIF":4.4,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11853615/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143482341","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}