Pub Date : 2025-12-01Epub Date: 2025-11-17DOI: 10.1007/s13402-025-01112-2
Daniel Tovar Manzano, Nabil Subhi-Issa, Alejandro Pereiro-Rodríguez, Igor Gregorio López Cade, María Mateos González, Miguel Fernández Arquero, Pedro Pérez Segura, Cristina Ujaldón Miró, Silvia Sánchez-Ramón, Alberto Ocaña, María Guzmán Fulgencio
{"title":"Tumor-infiltrating lymphocyte expansion protocols for adoptive cell therapy in cancer.","authors":"Daniel Tovar Manzano, Nabil Subhi-Issa, Alejandro Pereiro-Rodríguez, Igor Gregorio López Cade, María Mateos González, Miguel Fernández Arquero, Pedro Pérez Segura, Cristina Ujaldón Miró, Silvia Sánchez-Ramón, Alberto Ocaña, María Guzmán Fulgencio","doi":"10.1007/s13402-025-01112-2","DOIUrl":"10.1007/s13402-025-01112-2","url":null,"abstract":"","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"1659-1675"},"PeriodicalIF":4.8,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12698757/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145539302","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Spinal ependymal tumors are a diverse group of neoplasms encompassing four subtypes: spinal ependymoma (SP-EPN), spinal ependymoma, MYCN-amplification (SP-EPN-MYCN), spinal myxopapillary ependymoma (SP-MPE), and spinal subependymoma (SP-SE). However, the molecular differences among these subtypes remain largely unknown.
Methods: Using an integrated multi-omics approach (whole-genome sequencing, RNA-seq, and mass spectrometry), we identified the distinct molecular characteristics of three subtypes except for SP-EPN-MYCN.
Results: In SP-EPN, abnormal enrichment of ciliary signaling, particularly involving the MKS complex, was evident. SP-MPE exhibited significant dysregulation of mitochondrial metabolism, reflecting a metabolic profile aligned with the Warburg effect. SP-SE tumors showed enhanced activity of immune-related pathways, including interferon signaling and extracellular vesicle dynamics, suggesting a distinct tumor microenvironment.
Conclusion: This pilot study identifies candidate molecular markers in a single-center spinal ependymal tumor cohort.
{"title":"Multi-omics analysis delineates molecular signatures of spinal ependymal tumor.","authors":"Weihao Liu, Chao Ning, Xiaohan Geng, Bo Wang, Yaowu Zhang, Chong Wang, Yixiang Liu, Guanghao Zheng, Yongzhi Wang, Xinyu Wang, Dong Li, Wenqing Jia","doi":"10.1007/s13402-025-01122-0","DOIUrl":"10.1007/s13402-025-01122-0","url":null,"abstract":"<p><strong>Background: </strong>Spinal ependymal tumors are a diverse group of neoplasms encompassing four subtypes: spinal ependymoma (SP-EPN), spinal ependymoma, MYCN-amplification (SP-EPN-MYCN), spinal myxopapillary ependymoma (SP-MPE), and spinal subependymoma (SP-SE). However, the molecular differences among these subtypes remain largely unknown.</p><p><strong>Methods: </strong>Using an integrated multi-omics approach (whole-genome sequencing, RNA-seq, and mass spectrometry), we identified the distinct molecular characteristics of three subtypes except for SP-EPN-MYCN.</p><p><strong>Results: </strong>In SP-EPN, abnormal enrichment of ciliary signaling, particularly involving the MKS complex, was evident. SP-MPE exhibited significant dysregulation of mitochondrial metabolism, reflecting a metabolic profile aligned with the Warburg effect. SP-SE tumors showed enhanced activity of immune-related pathways, including interferon signaling and extracellular vesicle dynamics, suggesting a distinct tumor microenvironment.</p><p><strong>Conclusion: </strong>This pilot study identifies candidate molecular markers in a single-center spinal ependymal tumor cohort.</p><p><strong>Clinical trial number: </strong>Not applicable.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"1987-2000"},"PeriodicalIF":4.8,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12698791/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145399947","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-11-04DOI: 10.1007/s13402-025-01119-9
Kexin Yu, Yuanxiang Jin, Yidong Zhou, Qiaoping Xu
Hepatocellular carcinoma (HCC) remains one of the most lethal malignancies worldwide, with its pathogenesis involving complex biological processes such as DNA damage, epigenetic modification and oncogene mutation. Over the past two decades, the role of long non-coding RNAs (lncRNAs) in the occurrence, metastasis and progression of HCC has received increasing attention. As an important noncoding RNA molecule, lncRNAs play a key role in regulating gene expression, affecting RNA transcription and mRNA stability. This review elucidates the potential pathogenic processes of HCC and elaborates on the synthesis mechanisms of the above three non-coding RNAs. It comprehensively summarizes various non-coding RNAs that have been identified as playing key regulatory roles in HCC, as well as how these non-coding RNAs affect disease progression by regulating gene expression and protein functions. For example: lncRNAs such as NEAT1, DSCR8, PNUTS, HULC, and HOTAIR can play different roles in the proliferation, migration, and apoptosis of HCC cells in different ways. lncRNAs such as HClnc1, LINC01343, FAM111A-DT, CERS6-AS1, and TLNC1 significantly affect the progression of HCC by regulating key signaling axes or protein functions, and are closely related to the prognosis of patients. In addition, we also discuss the potential of lncRNAs as therapeutic targets for HCC, such as: lncRNA MIR31HG, CASC2c, and lncRNA AC115619. Furthermore, we also explore the application prospects of lncRNAs as potential biomarkers and therapeutic targets, providing new perspectives and directions for future HCC research.
{"title":"Long non-coding RNA in hepatocellular carcinoma: mechanistic insights and therapeutic perspectives.","authors":"Kexin Yu, Yuanxiang Jin, Yidong Zhou, Qiaoping Xu","doi":"10.1007/s13402-025-01119-9","DOIUrl":"10.1007/s13402-025-01119-9","url":null,"abstract":"<p><p>Hepatocellular carcinoma (HCC) remains one of the most lethal malignancies worldwide, with its pathogenesis involving complex biological processes such as DNA damage, epigenetic modification and oncogene mutation. Over the past two decades, the role of long non-coding RNAs (lncRNAs) in the occurrence, metastasis and progression of HCC has received increasing attention. As an important noncoding RNA molecule, lncRNAs play a key role in regulating gene expression, affecting RNA transcription and mRNA stability. This review elucidates the potential pathogenic processes of HCC and elaborates on the synthesis mechanisms of the above three non-coding RNAs. It comprehensively summarizes various non-coding RNAs that have been identified as playing key regulatory roles in HCC, as well as how these non-coding RNAs affect disease progression by regulating gene expression and protein functions. For example: lncRNAs such as NEAT1, DSCR8, PNUTS, HULC, and HOTAIR can play different roles in the proliferation, migration, and apoptosis of HCC cells in different ways. lncRNAs such as HClnc1, LINC01343, FAM111A-DT, CERS6-AS1, and TLNC1 significantly affect the progression of HCC by regulating key signaling axes or protein functions, and are closely related to the prognosis of patients. In addition, we also discuss the potential of lncRNAs as therapeutic targets for HCC, such as: lncRNA MIR31HG, CASC2c, and lncRNA AC115619. Furthermore, we also explore the application prospects of lncRNAs as potential biomarkers and therapeutic targets, providing new perspectives and directions for future HCC research.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"1677-1692"},"PeriodicalIF":4.8,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12698749/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145437242","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Purpose: Combining paclitaxel with antiangiogenic agents has demonstrated improved efficacy as second-line treatment for advanced gastric cancer. Surufatinib, a multi-kinase inhibitor with antiangiogenic and immunomodulatory properties, has exhibited synergistic effects with chemotherapy in preclinical studies.
Methods: This single-arm phase 2 trial enrolled patients aged 18-75 with HER2-negative unresectable or metastatic gastric/gastroesophageal junction adenocarcinoma who had failed first-line therapy. All received surufatinib 250 mg once daily plus paclitaxel 150mg/m2 every 3 weeks for up to 6 cycles, followed by maintenance surufatinib until progression, intolerable toxicity, or withdrawal. The primary endpoint was objective response rate (ORR) per Response Evaluation Criteria in Solid Tumors version 1.1. Secondary endpoints included disease control rate (DCR), progression-free survival (PFS), overall survival (OS), and safety.
Results: Thirty-five patients were enrolled. Among 32 tumor response-evaluable patients, the ORR and DCR were 25.0% (95% confidence interval [CI]: 11.5, 43.4) and 87.5% (95% CI: 71.0, 96.5), respectively. Median PFS was 5.7 (95% CI: 4.7, 6.9) months and median OS was 10.8 (95% CI: 7.0, 17.2) months. In 26 patients with prior immunotherapy exposure, the median OS was 14.4 (95% CI: 8.5, not estimable) months. Overall, treatment-related adverse events of grade ≥ 3 occurred in 19 (54.3%) patients, with neutropenia (40.0%), leukopenia (34.3%), and hypertension (11.4%) being the most commonly observed.
Conclusions: Surufatinib plus paclitaxel showed promising efficacy and manageable safety as second-line treatment for advanced gastric cancer, especially in patients who had failed prior immunotherapy.
Clinical trial number: ChiCTR2200063336, registered in the Chinese Clinical Trial Registry on September 5, 2022.
{"title":"Surufatinib plus paclitaxel as second-line therapy for advanced gastric cancer: a single-arm, phase 2 clinical trial.","authors":"Ting Han, Meng Zhuo, Xinyi Zheng, Xiuqi Wu, Feng Jiao, Jiujie Cui, Tao Wang, Xiaoxia Qiu, Yingjie Su, Xiaolin Lin, Xiuying Xiao","doi":"10.1007/s13402-025-01127-9","DOIUrl":"10.1007/s13402-025-01127-9","url":null,"abstract":"<p><strong>Purpose: </strong>Combining paclitaxel with antiangiogenic agents has demonstrated improved efficacy as second-line treatment for advanced gastric cancer. Surufatinib, a multi-kinase inhibitor with antiangiogenic and immunomodulatory properties, has exhibited synergistic effects with chemotherapy in preclinical studies.</p><p><strong>Methods: </strong>This single-arm phase 2 trial enrolled patients aged 18-75 with HER2-negative unresectable or metastatic gastric/gastroesophageal junction adenocarcinoma who had failed first-line therapy. All received surufatinib 250 mg once daily plus paclitaxel 150mg/m<sup>2</sup> every 3 weeks for up to 6 cycles, followed by maintenance surufatinib until progression, intolerable toxicity, or withdrawal. The primary endpoint was objective response rate (ORR) per Response Evaluation Criteria in Solid Tumors version 1.1. Secondary endpoints included disease control rate (DCR), progression-free survival (PFS), overall survival (OS), and safety.</p><p><strong>Results: </strong>Thirty-five patients were enrolled. Among 32 tumor response-evaluable patients, the ORR and DCR were 25.0% (95% confidence interval [CI]: 11.5, 43.4) and 87.5% (95% CI: 71.0, 96.5), respectively. Median PFS was 5.7 (95% CI: 4.7, 6.9) months and median OS was 10.8 (95% CI: 7.0, 17.2) months. In 26 patients with prior immunotherapy exposure, the median OS was 14.4 (95% CI: 8.5, not estimable) months. Overall, treatment-related adverse events of grade ≥ 3 occurred in 19 (54.3%) patients, with neutropenia (40.0%), leukopenia (34.3%), and hypertension (11.4%) being the most commonly observed.</p><p><strong>Conclusions: </strong>Surufatinib plus paclitaxel showed promising efficacy and manageable safety as second-line treatment for advanced gastric cancer, especially in patients who had failed prior immunotherapy.</p><p><strong>Clinical trial number: </strong>ChiCTR2200063336, registered in the Chinese Clinical Trial Registry on September 5, 2022.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"2073-2084"},"PeriodicalIF":4.8,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12698750/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145437215","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Purpose: Tumor cells reprogram their fatty acid metabolism to meet the demands for their rapid proliferation. However, the interplay between fatty acid metabolism and the tumor microenvironment (TME) in lung cancer remains poorly defined. This study aims to elucidate how arachidonic acid (AA) metabolism, specifically via the enzyme 5-lipoxygenase (ALOX5), modulates anti-tumor immunity in non-small cell lung cancer (NSCLC).
Methods: Data from public transcriptomic datasets were analyzed to identify differentially expressed and immune regulatory fatty acid metabolism-related genes in NSCLC. Spatial correlation between ALOX5 expression and CD8⁺ T cell infiltration was assessed via immunofluorescence. Functional impacts of ALOX5 on tumor growth, immune recruitment, and immunotherapy response were characterized using knockdown and overexpression models. Clinical relevance was evaluated by profiling plasma fatty acids via mass spectrometry in immunotherapy-treated cohorts.
Results: Bioinformatic analysis nominated ALOX5-mediatedAA metabolic pathway as a key regulator of immune infiltration. Genetic knockdown of ALOX5 accelerated tumor progression, attenuated CD8⁺ T cell recruitment, and reduced leukotriene B4 (LTB4) production alongside downregulation of cytotoxic (granzymes) and chemotactic genes. Conversely, ALOX5 overexpression suppressed tumor growth and synergized with anti-PD-1 therapy. Exogenous AA supplementation similarly potentiated the efficacy of PD-1 blockade in vivo. Clinically, elevated plasma levels of AA and linoleic acid correlated with improved immunotherapy response and survival outcomes.
Conclusion: Tumor-intrinsic ALOX5 is a novel tumor suppressor that orchestrates CD8⁺ T infiltration via the AA-LTB4 axis in NSCLC. Our findings establish ALOX5-mediated AA metabolism as a therapeutically targetable pathway to overcome immunotherapy resistance, positioning dietary AA supplementation as a promising adjunctive strategy.
{"title":"Tumoral ALOX5 mediated arachidonic acid metabolism regulates immune response in non-small cell lung cancer.","authors":"Yuan Gao, Yangchen Xia, Jiayao Li, Yongfeng Lai, Songlin Yin, Junhong Lin, Dongrunhan Yu, Shanshan Huang, Fujia Lu, Weimin Wang, Qian Chu","doi":"10.1007/s13402-025-01124-y","DOIUrl":"10.1007/s13402-025-01124-y","url":null,"abstract":"<p><strong>Purpose: </strong>Tumor cells reprogram their fatty acid metabolism to meet the demands for their rapid proliferation. However, the interplay between fatty acid metabolism and the tumor microenvironment (TME) in lung cancer remains poorly defined. This study aims to elucidate how arachidonic acid (AA) metabolism, specifically via the enzyme 5-lipoxygenase (ALOX5), modulates anti-tumor immunity in non-small cell lung cancer (NSCLC).</p><p><strong>Methods: </strong>Data from public transcriptomic datasets were analyzed to identify differentially expressed and immune regulatory fatty acid metabolism-related genes in NSCLC. Spatial correlation between ALOX5 expression and CD8⁺ T cell infiltration was assessed via immunofluorescence. Functional impacts of ALOX5 on tumor growth, immune recruitment, and immunotherapy response were characterized using knockdown and overexpression models. Clinical relevance was evaluated by profiling plasma fatty acids via mass spectrometry in immunotherapy-treated cohorts.</p><p><strong>Results: </strong>Bioinformatic analysis nominated ALOX5-mediatedAA metabolic pathway as a key regulator of immune infiltration. Genetic knockdown of ALOX5 accelerated tumor progression, attenuated CD8⁺ T cell recruitment, and reduced leukotriene B4 (LTB4) production alongside downregulation of cytotoxic (granzymes) and chemotactic genes. Conversely, ALOX5 overexpression suppressed tumor growth and synergized with anti-PD-1 therapy. Exogenous AA supplementation similarly potentiated the efficacy of PD-1 blockade in vivo. Clinically, elevated plasma levels of AA and linoleic acid correlated with improved immunotherapy response and survival outcomes.</p><p><strong>Conclusion: </strong>Tumor-intrinsic ALOX5 is a novel tumor suppressor that orchestrates CD8⁺ T infiltration via the AA-LTB4 axis in NSCLC. Our findings establish ALOX5-mediated AA metabolism as a therapeutically targetable pathway to overcome immunotherapy resistance, positioning dietary AA supplementation as a promising adjunctive strategy.</p><p><strong>Clinical trail number: </strong>Not applicable.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"2017-2034"},"PeriodicalIF":4.8,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12698827/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145444256","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-11-20DOI: 10.1007/s13402-025-01104-2
Chenhuan Ding, Chen Wang, Junfeng Guo, Yi Lai, Yingbin Wang, He Li
Background: Despite advances in ovarian cancer treatment, the tendency for cancer cells to metastasise to the peritoneum still results in poor prognosis. Studies have demonstrated that the integrin family plays a role in this metastasis; however, the underlying mechanism remains unclear. Triptolide (TP) has been confirmed to have a strong cytotoxic effect against ovarian cancer. However, its clinical application is limited by its severe systemic toxicity and low water solubility.
Methods: This study investigated the integrins involved in peritoneal metastasis and their associated mechanisms. Furthermore, Si/TP@Exos were constructed to counteract the metastatic potential of ovarian cancer cells.
Results: In vitro experiments showed that the construction of the ITGA4B2/AEP ternary complex contributed to the peritoneal metastasis of ovarian cancer by activating the IL-17 and NF-kappa B signalling pathways. Thus, whether the combined application of siRNA targeting ITGA4B2 and TP could further overcome peritoneal metastasis in ovarian cancer was investigated. In vitro results indicated that Si/TP@Exos were efficiently taken up by ovarian cancer cells, thus significantly enhancing the apoptosis of tumor cells. Similarly, Si/TP@Exos were effectively enriched in the tumor areas and exerted anti-tumor activity obviously in vivo.
Conclusions: Together, these findings present a novel strategy to overcome the peritoneal metastasis tendency of ovarian cancer and offer a potential therapeutic solution for clinical treatment of ovarian cancer. The combination of traditional Chinese medicine nano drug delivery platforms provides a new perspective for cancer treatment.
{"title":"A Tumor-homing nanoplatform for the co-delivery of triptolide and siRNA-A4B2 conspicuously overcomes peritoneum metastasis of ovarian cancer.","authors":"Chenhuan Ding, Chen Wang, Junfeng Guo, Yi Lai, Yingbin Wang, He Li","doi":"10.1007/s13402-025-01104-2","DOIUrl":"10.1007/s13402-025-01104-2","url":null,"abstract":"<p><strong>Background: </strong>Despite advances in ovarian cancer treatment, the tendency for cancer cells to metastasise to the peritoneum still results in poor prognosis. Studies have demonstrated that the integrin family plays a role in this metastasis; however, the underlying mechanism remains unclear. Triptolide (TP) has been confirmed to have a strong cytotoxic effect against ovarian cancer. However, its clinical application is limited by its severe systemic toxicity and low water solubility.</p><p><strong>Methods: </strong>This study investigated the integrins involved in peritoneal metastasis and their associated mechanisms. Furthermore, Si/TP@Exos were constructed to counteract the metastatic potential of ovarian cancer cells.</p><p><strong>Results: </strong>In vitro experiments showed that the construction of the ITGA4B2/AEP ternary complex contributed to the peritoneal metastasis of ovarian cancer by activating the IL-17 and NF-kappa B signalling pathways. Thus, whether the combined application of siRNA targeting ITGA4B2 and TP could further overcome peritoneal metastasis in ovarian cancer was investigated. In vitro results indicated that Si/TP@Exos were efficiently taken up by ovarian cancer cells, thus significantly enhancing the apoptosis of tumor cells. Similarly, Si/TP@Exos were effectively enriched in the tumor areas and exerted anti-tumor activity obviously in vivo.</p><p><strong>Conclusions: </strong>Together, these findings present a novel strategy to overcome the peritoneal metastasis tendency of ovarian cancer and offer a potential therapeutic solution for clinical treatment of ovarian cancer. The combination of traditional Chinese medicine nano drug delivery platforms provides a new perspective for cancer treatment.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"1741-1756"},"PeriodicalIF":4.8,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12698833/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145562803","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Purpose: Cancer-associated fibroblasts (CAFs) are major constituents of the tumor microenvironment (TME) and have been associated with chemotherapeutic failure via different mechanisms. However, the CAFs inhibit chemotherapy mechanism in lung adenocarcinoma (LUAD) remains undetermined.
Methods: Fibroblasts were isolated from tumor and normal lung tissues from patients with poorly differentiated LUAD (pCAFs), moderately differentiated LUAD (mCAFs), and normal fibroblasts (NFs). Then, the influence of these fibroblasts on carboplatin's cytotoxic effects on LUAD cell lines A549 and NCI-H1299 was assessed by measuring their IC50 values. Furthermore, CXCL12 secretion and its role in chemotherapeutics were also evaluated.
Results: The data revealed that pCAFs significantly inhibited apoptosis in LUAD cells and increased carboplatin IC50 values. Furthermore, pCAFs secreted higher CXCL12 content than mCAFs and NFs. Moreover, in pCAFs, CXCL12 silencing enhanced carboplatin's cytotoxic effects, while NFs overexpressing CXCL12 inhibited carboplatin's efficacy. Mechanistically, pCAFs promote the secretion of CXCL12 by activating the NF-κB pathway, and CXCL12 binds to CXCR4 on LUAD cells, thereby promoting carboplatin resistance. Moreover, in the xenograft models, pCAFs were found to reduce carboplatin's cytotoxicity by CXCL12 secretion. Moreover, the analysis of the LUAD patient's tumor and peripheral blood sample indicated a correlation between lower differentiation and higher CXCL12 expression levels.
Conclusion: This study revealed that LUAD-derived CAFs activate the NF-κB axis to secrete CXCL12, thereby weakening the carboplatin's killing effect on LUAD. Furthermore, poorly differentiated LUAD secreted more CXCL12. These findings indicate a novel strategy to enhance carboplatin's chemotherapeutic potential against LUAD.
{"title":"CAFs mediate carboplatin resistance in LUAD via CXCL12 secretion regulated by NF-κB activation.","authors":"Long Li, Xu Zhu, Zeyang Yang, Xuanyin Wang, Xianling Zeng, Lu Wang, Jiaming Ren, Jiangwei Wu, Qiaoling Zhang, Jing Xiao, Ying Zhang, Song Yang, Yuanhan Wang, Jian Zhang, Xinlei Liu, Siyuan Yang, Zhu Zeng, Jieheng Wu","doi":"10.1007/s13402-025-01106-0","DOIUrl":"10.1007/s13402-025-01106-0","url":null,"abstract":"<p><strong>Purpose: </strong>Cancer-associated fibroblasts (CAFs) are major constituents of the tumor microenvironment (TME) and have been associated with chemotherapeutic failure via different mechanisms. However, the CAFs inhibit chemotherapy mechanism in lung adenocarcinoma (LUAD) remains undetermined.</p><p><strong>Methods: </strong>Fibroblasts were isolated from tumor and normal lung tissues from patients with poorly differentiated LUAD (pCAFs), moderately differentiated LUAD (mCAFs), and normal fibroblasts (NFs). Then, the influence of these fibroblasts on carboplatin's cytotoxic effects on LUAD cell lines A549 and NCI-H1299 was assessed by measuring their IC<sub>50</sub> values. Furthermore, CXCL12 secretion and its role in chemotherapeutics were also evaluated.</p><p><strong>Results: </strong>The data revealed that pCAFs significantly inhibited apoptosis in LUAD cells and increased carboplatin IC<sub>50</sub> values. Furthermore, pCAFs secreted higher CXCL12 content than mCAFs and NFs. Moreover, in pCAFs, CXCL12 silencing enhanced carboplatin's cytotoxic effects, while NFs overexpressing CXCL12 inhibited carboplatin's efficacy. Mechanistically, pCAFs promote the secretion of CXCL12 by activating the NF-κB pathway, and CXCL12 binds to CXCR4 on LUAD cells, thereby promoting carboplatin resistance. Moreover, in the xenograft models, pCAFs were found to reduce carboplatin's cytotoxicity by CXCL12 secretion. Moreover, the analysis of the LUAD patient's tumor and peripheral blood sample indicated a correlation between lower differentiation and higher CXCL12 expression levels.</p><p><strong>Conclusion: </strong>This study revealed that LUAD-derived CAFs activate the NF-κB axis to secrete CXCL12, thereby weakening the carboplatin's killing effect on LUAD. Furthermore, poorly differentiated LUAD secreted more CXCL12. These findings indicate a novel strategy to enhance carboplatin's chemotherapeutic potential against LUAD.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"1775-1794"},"PeriodicalIF":4.8,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12698753/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145205797","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Purpose: Natural killer (NK) cells mediate anti-tumor immunity through integrated signaling of inhibitory and activating receptors. The efficacy of NK cell adoptive transfer therapy varies among patients due to heterogeneous receptor-ligand expression. This study aimed to develop a predictive model based on receptor-ligand interactions to determine NK cells' therapeutic effects.
Methods: Through analyses of receptor-ligand expression profiles of NK and tumor cells and assessment of NK cell cytotoxicity, we developed a machine learning-based random forest model using 11 key receptor-ligand pairs selected through database mining and experimental screening. Flow cytometry was used to obtain receptor-ligand profiles, and combined predictors were calculated for each pair. The model was validated using independent datasets and evaluated for generalizability across different tumor types.
Results: The model showed significant predictive performance, achieving an accuracy of 84.2% and an area under the curve (AUC) of 0.908 in ovarian cancer cohorts. This predictive capability was validated in both in vitro experiments and clinical samples, revealing complex non-linear interactions between receptor-ligand expression and NK cell killing efficacy. Cancer-specific ligand expression patterns were identified. While showing optimal performance in studied cancer types, it exhibited moderate applicability to other cancers and demonstrated potential compatibility with transcriptomic data for prediction.
Conclusions: This model provides tools and foundations for the precise treatment of tumors using NK immune cells and may be applied in clinical practice.
{"title":"Machine learning-based development of a cytotoxicity prediction model for NK cell therapy in cancers.","authors":"Jie Ma, Jingjing Yue, Yangyang Li, Yutong Li, Hongbo Dong, Fang Fang, Weihua Xiao","doi":"10.1007/s13402-025-01113-1","DOIUrl":"10.1007/s13402-025-01113-1","url":null,"abstract":"<p><strong>Purpose: </strong>Natural killer (NK) cells mediate anti-tumor immunity through integrated signaling of inhibitory and activating receptors. The efficacy of NK cell adoptive transfer therapy varies among patients due to heterogeneous receptor-ligand expression. This study aimed to develop a predictive model based on receptor-ligand interactions to determine NK cells' therapeutic effects.</p><p><strong>Methods: </strong>Through analyses of receptor-ligand expression profiles of NK and tumor cells and assessment of NK cell cytotoxicity, we developed a machine learning-based random forest model using 11 key receptor-ligand pairs selected through database mining and experimental screening. Flow cytometry was used to obtain receptor-ligand profiles, and combined predictors were calculated for each pair. The model was validated using independent datasets and evaluated for generalizability across different tumor types.</p><p><strong>Results: </strong>The model showed significant predictive performance, achieving an accuracy of 84.2% and an area under the curve (AUC) of 0.908 in ovarian cancer cohorts. This predictive capability was validated in both in vitro experiments and clinical samples, revealing complex non-linear interactions between receptor-ligand expression and NK cell killing efficacy. Cancer-specific ligand expression patterns were identified. While showing optimal performance in studied cancer types, it exhibited moderate applicability to other cancers and demonstrated potential compatibility with transcriptomic data for prediction.</p><p><strong>Conclusions: </strong>This model provides tools and foundations for the precise treatment of tumors using NK immune cells and may be applied in clinical practice.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"1837-1870"},"PeriodicalIF":4.8,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12698772/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145421251","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-10-29DOI: 10.1007/s13402-025-01120-2
Man Zhang, Caixia Chen, Yuekun Fang, Wanying Liu, Xiaoying Zhang, Liting Chen, Yi Xiao
Background: Chimeric antigen receptor (CAR) T cells have long been regarded as living drugs, those activation, diffusion, and expansion influence the clinical efficacy. Dynamic monitoring of CAR-T cells in vivo is crucial for understanding their biodistribution and optimizing therapeutic outcomes. However, due to the challenges in sampling tissue-resident CAR-T cells from patients, limited data have been reported on their spatial and temporal distribution.
Methods: This study enrolled 43 patients with haematological malignancies receiving CAR-T cell therapy. CAR copy numbers in the peripheral blood (PB) and serous cavity effusion (SCE) were sequentially quantified and analysed.
Results: High expansion of CAR-T cells in PB was associated with subsequent expansion in SCE. The median Tmax in SCE occurred later than in PB. Patients with immune effector cell-associated neurotoxicity syndrome (ICANS) exhibited higher CAR copy numbers in cerebrospinal fluid (CSF) compared to those without ICANS. Peripheral infection was associated with increased CAR copy numbers in CSF, which may be caused by cell diffusion or/and expansion. Tumour invasion favored local accumulation and expansion of CAR-T cells in pleural effusion or ascites (PE/A), and patients with tumour invasion had a higher incidence of local cytokine release syndrome (L-CRS).
Conclusion: We characterized the spatial and temporal distribution of CAR-T cells and identified associations between CAR copy numbers and local inflammation, tumour invasion, and adverse events. These findings enhance our understanding of CAR-T cells diffusion, trafficking, and expansion, providing novel insights for clinical management and therapeutic optimization.
{"title":"Spatio-temporal kinetics of patients' CAR-T cells in blood and serous cavity effusion: real-world clinical insights.","authors":"Man Zhang, Caixia Chen, Yuekun Fang, Wanying Liu, Xiaoying Zhang, Liting Chen, Yi Xiao","doi":"10.1007/s13402-025-01120-2","DOIUrl":"10.1007/s13402-025-01120-2","url":null,"abstract":"<p><strong>Background: </strong>Chimeric antigen receptor (CAR) T cells have long been regarded as living drugs, those activation, diffusion, and expansion influence the clinical efficacy. Dynamic monitoring of CAR-T cells in vivo is crucial for understanding their biodistribution and optimizing therapeutic outcomes. However, due to the challenges in sampling tissue-resident CAR-T cells from patients, limited data have been reported on their spatial and temporal distribution.</p><p><strong>Methods: </strong>This study enrolled 43 patients with haematological malignancies receiving CAR-T cell therapy. CAR copy numbers in the peripheral blood (PB) and serous cavity effusion (SCE) were sequentially quantified and analysed.</p><p><strong>Results: </strong>High expansion of CAR-T cells in PB was associated with subsequent expansion in SCE. The median T<sub>max</sub> in SCE occurred later than in PB. Patients with immune effector cell-associated neurotoxicity syndrome (ICANS) exhibited higher CAR copy numbers in cerebrospinal fluid (CSF) compared to those without ICANS. Peripheral infection was associated with increased CAR copy numbers in CSF, which may be caused by cell diffusion or/and expansion. Tumour invasion favored local accumulation and expansion of CAR-T cells in pleural effusion or ascites (PE/A), and patients with tumour invasion had a higher incidence of local cytokine release syndrome (L-CRS).</p><p><strong>Conclusion: </strong>We characterized the spatial and temporal distribution of CAR-T cells and identified associations between CAR copy numbers and local inflammation, tumour invasion, and adverse events. These findings enhance our understanding of CAR-T cells diffusion, trafficking, and expansion, providing novel insights for clinical management and therapeutic optimization.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"1955-1969"},"PeriodicalIF":4.8,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12698831/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145399960","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}