Pengrui Cheng, Que Yang, Xiao Zhang, Qiang Wang, Bingzheng Zhong
DNA-dependent activator of interferon-regulatory factors (DAI) has recently been identified to trigger ferroptosis in endothelial cells. However, it remains unclear whether it can also elicit ferroptosis in tumor cells and further remodel the tumor immune microenvironment (TIME). In this study, we found that activation of DAI could also trigger mouse colorectal cancer (CRC) cells ferroptosis. Further experiments showed that DAI-driven ferroptosis induced mitochondria oxidative stress and dysfunction, leading to the release of mitochondrial DNA (mtDNA) into the cytoplasm, which subsequently activated the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway and thereby reprogrammed the TIME by promoting tumor-associated macrophages (TAMs) M1 polarization while preventing TAMs from polarizing towards M2 type, exerting an effective anti-tumor effect, which significantly reduced tumor size and weight. In summary, our findings confirmed DAI-triggered ferroptosis-induced mtDNA-mediated cGAS-STING anti-tumor immunity pathway in mouse CRC cells, providing novel insights into the development of more effective tumor immunotherapeutic strategies that are based on DAI-mediated programmed cell death (PCD).
{"title":"Activation of cGAS-STING Pathway by DAI-Triggered Ferroptosis in CRC Cells Reprograms TAMs Balance to Promote Anti-Tumor Immunity","authors":"Pengrui Cheng, Que Yang, Xiao Zhang, Qiang Wang, Bingzheng Zhong","doi":"10.1111/cas.70196","DOIUrl":"10.1111/cas.70196","url":null,"abstract":"<p>DNA-dependent activator of interferon-regulatory factors (DAI) has recently been identified to trigger ferroptosis in endothelial cells. However, it remains unclear whether it can also elicit ferroptosis in tumor cells and further remodel the tumor immune microenvironment (TIME). In this study, we found that activation of DAI could also trigger mouse colorectal cancer (CRC) cells ferroptosis. Further experiments showed that DAI-driven ferroptosis induced mitochondria oxidative stress and dysfunction, leading to the release of mitochondrial DNA (mtDNA) into the cytoplasm, which subsequently activated the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway and thereby reprogrammed the TIME by promoting tumor-associated macrophages (TAMs) M1 polarization while preventing TAMs from polarizing towards M2 type, exerting an effective anti-tumor effect, which significantly reduced tumor size and weight. In summary, our findings confirmed DAI-triggered ferroptosis-induced mtDNA-mediated cGAS-STING anti-tumor immunity pathway in mouse CRC cells, providing novel insights into the development of more effective tumor immunotherapeutic strategies that are based on DAI-mediated programmed cell death (PCD).</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"116 12","pages":"3286-3299"},"PeriodicalIF":4.3,"publicationDate":"2025-09-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cas.70196","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145193608","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}
While the development of innovative drugs has improved the outcomes of various cancers in recent years, the number of patients with oral squamous cell carcinoma (OSCC) and mortality rates has not yet decreased significantly. This could be attributed to the prognostic and molecular differences occurring at different locations of cancer development. In this study, we conducted gene expression, DNA methylation, and tumor immunological analyses of gingival squamous cell carcinoma (GSCC) and tongue squamous cell carcinoma (TSCC) to elucidate the characteristic gene expression changes, their mechanisms, and tumor immune profiles. Gene expression analysis suggested that pathways related to the cell cycle and antibacterial humoral immunity were upregulated in GSCC, whereas immune response pathways were upregulated in TSCC. Additionally, high HOXC13 expression may be associated with GSCC prognosis. DNA methylation analysis revealed hypermethylation of the 3′ UTR of the HOXD11 gene, leading to increased expression in both GSCC and TSCC. In the tumor immune profile analysis, when comparing tumor and normal tissues in GSCC, the number of immune cells did not change significantly; however, the proportion of inflammatory cells, such as CD8+ T cells, changed. In TSCC, the number and percentage of inflammatory cells, such as T lymphocytes, increased in the tumor compared to those of normal tissues. These findings underscore the importance of understanding the characteristics of site-specific OSCC. Therefore, it is essential to establish standardized treatment protocols and develop novel therapeutic strategies tailored to each anatomical site of the tumor.
{"title":"Variation of Gene Expression and Methylation Profiling in Gingival and Tongue Cancers","authors":"Takafumi Kashiwagi, So Takata, Hidenori Tanaka, Shoko Ikuta, Yasushi Totoki, Yoichiro Nakatani, Hidenori Inohara, Narikazu Uzawa, Shinichi Yachida","doi":"10.1111/cas.70205","DOIUrl":"10.1111/cas.70205","url":null,"abstract":"<p>While the development of innovative drugs has improved the outcomes of various cancers in recent years, the number of patients with oral squamous cell carcinoma (OSCC) and mortality rates has not yet decreased significantly. This could be attributed to the prognostic and molecular differences occurring at different locations of cancer development. In this study, we conducted gene expression, DNA methylation, and tumor immunological analyses of gingival squamous cell carcinoma (GSCC) and tongue squamous cell carcinoma (TSCC) to elucidate the characteristic gene expression changes, their mechanisms, and tumor immune profiles. Gene expression analysis suggested that pathways related to the cell cycle and antibacterial humoral immunity were upregulated in GSCC, whereas immune response pathways were upregulated in TSCC. Additionally, high <i>HOXC13</i> expression may be associated with GSCC prognosis. DNA methylation analysis revealed hypermethylation of the 3′ UTR of the <i>HOXD11</i> gene, leading to increased expression in both GSCC and TSCC. In the tumor immune profile analysis, when comparing tumor and normal tissues in GSCC, the number of immune cells did not change significantly; however, the proportion of inflammatory cells, such as CD8+ T cells, changed. In TSCC, the number and percentage of inflammatory cells, such as T lymphocytes, increased in the tumor compared to those of normal tissues. These findings underscore the importance of understanding the characteristics of site-specific OSCC. Therefore, it is essential to establish standardized treatment protocols and develop novel therapeutic strategies tailored to each anatomical site of the tumor.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"116 12","pages":"3459-3472"},"PeriodicalIF":4.3,"publicationDate":"2025-09-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cas.70205","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145187063","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}
Qian Li, Xu Huang, Yan Tong, Caiyu Yong, Minghui Song, Cunjie Chang, Heng Dong, Fangtian Bu, Shuanghong Yan, Jie Ying, Jianxiang Chen
circRNAs are endogenous covalently closed circular RNAs, and previous studies have validated their key regulatory roles in malignant tumors; however, the specific role and regulatory mechanism of circNEK11 in hepatocellular carcinoma (HCC) remain unclear. Here, we discovered that circNEK11 back-spliced from exons 12, 13, 14, and 15 of the NEK11 gene is significantly upregulated in HCC tissues, especially recurrent HCC tissues. YTHDC1 functions as a reader of m6A-modified circNEK11 and facilitates its cytoplasmic export. circNEK11 promotes HCC cell oncogenic functions in vitro and HCC growth in vivo. Mechanistically, circNEK11 directly binds to miR-1236-3p to regulate GPX2 expression, thus promoting HCC progression and recurrence. Collectively, our findings suggest that circNEK11 is a potential biomarker and target for HCC diagnosis and treatment.
{"title":"N6-Methyladenosine-Modified circNEK11 Promotes Hepatocellular Carcinoma Progression via the miR-1236-3p/GPX2 Axis","authors":"Qian Li, Xu Huang, Yan Tong, Caiyu Yong, Minghui Song, Cunjie Chang, Heng Dong, Fangtian Bu, Shuanghong Yan, Jie Ying, Jianxiang Chen","doi":"10.1111/cas.70203","DOIUrl":"10.1111/cas.70203","url":null,"abstract":"<p>circRNAs are endogenous covalently closed circular RNAs, and previous studies have validated their key regulatory roles in malignant tumors; however, the specific role and regulatory mechanism of circNEK11 in hepatocellular carcinoma (HCC) remain unclear. Here, we discovered that circNEK11 back-spliced from exons 12, 13, 14, and 15 of the NEK11 gene is significantly upregulated in HCC tissues, especially recurrent HCC tissues. YTHDC1 functions as a reader of m6A-modified circNEK11 and facilitates its cytoplasmic export. circNEK11 promotes HCC cell oncogenic functions in vitro and HCC growth in vivo. Mechanistically, circNEK11 directly binds to miR-1236-3p to regulate GPX2 expression, thus promoting HCC progression and recurrence. Collectively, our findings suggest that circNEK11 is a potential biomarker and target for HCC diagnosis and treatment.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"116 12","pages":"3326-3336"},"PeriodicalIF":4.3,"publicationDate":"2025-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cas.70203","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145132433","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}
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of advanced renal cell carcinoma (RCC). However, ICIs often induce immune-related adverse events (irAEs), which vary greatly among individuals and may influence treatment outcomes. This study aimed to identify genetic markers associated with the risk of severe treatment-related adverse events (trAEs) and assess their impact on patient prognosis. From August 19, 2019, to September 30, 2020, patient recruitment for nivolumab treatment in advanced clear cell RCC (ccRCC) was conducted across 23 institutions in Japan, with follow-up concluding on March 31, 2021 (protocol ID: UMIN000037739). A genome-wide association study (GWAS) was conducted in a development cohort to identify single nucleotide polymorphisms (SNPs) associated with severe trAEs following nivolumab. Sixteen SNPs were identified, and thirteen were genotyped in a validation cohort. Eight SNPs showed consistent trends with the development cohort, but they have not reached statistical significance in the validation cohort. Among them, rs2545737, corresponding to CHD1, was significantly linked to prolonged progression-free survival (PFS), highlighting its potential as a biomarker for both safety and efficacy. Further analysis indicated that high CHD1 expression in tumors correlated with improved overall survival in nivolumab-treated patients but not in those receiving everolimus. Given the failure to replicate the development set findings in our validation cohort, further re-validation within the RCC population is warranted. However, these results enhance our understanding of the genetic predisposition to trAEs and provide a significant step toward safer and more effective cancer treatment strategies. This study was registered on the University Hospital Medical Information Network (UMIN) in Japan on August 20, 2019 (protocol ID: UMIN000037739).
{"title":"GWAS Identifies SNPs Associated With Severe Adverse Events and Efficacy in Advanced Renal Cell Carcinoma Treated With Nivolumab","authors":"Tokiyoshi Tanegashima, Masaki Shiota, Shusuke Akamatsu, Hideaki Miyake, Masayuki Takahashi, Mototsugu Oya, Norihiko Tsuchiya, Naoya Masumori, Hideyasu Matsuyama, Wataru Obara, Nobuo Shinohara, Kiyohide Fujimoto, Masahiro Nozawa, Kojiro Ohba, Chikara Ohyama, Katsuyoshi Hashine, Tomomi Kamba, Koji Mita, Momokazu Gotoh, Shuichi Tatarano, Masato Fujisawa, Yoshihiko Tomita, Shoichiro Mukai, Keiichi Ito, Shoji Tokunaga, Masatoshi Eto, SNiP-RCC investigators","doi":"10.1111/cas.70204","DOIUrl":"10.1111/cas.70204","url":null,"abstract":"<p>Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of advanced renal cell carcinoma (RCC). However, ICIs often induce immune-related adverse events (irAEs), which vary greatly among individuals and may influence treatment outcomes. This study aimed to identify genetic markers associated with the risk of severe treatment-related adverse events (trAEs) and assess their impact on patient prognosis. From August 19, 2019, to September 30, 2020, patient recruitment for nivolumab treatment in advanced clear cell RCC (ccRCC) was conducted across 23 institutions in Japan, with follow-up concluding on March 31, 2021 (protocol ID: UMIN000037739). A genome-wide association study (GWAS) was conducted in a development cohort to identify single nucleotide polymorphisms (SNPs) associated with severe trAEs following nivolumab. Sixteen SNPs were identified, and thirteen were genotyped in a validation cohort. Eight SNPs showed consistent trends with the development cohort, but they have not reached statistical significance in the validation cohort. Among them, rs2545737, corresponding to <i>CHD1</i>, was significantly linked to prolonged progression-free survival (PFS), highlighting its potential as a biomarker for both safety and efficacy. Further analysis indicated that high <i>CHD1</i> expression in tumors correlated with improved overall survival in nivolumab-treated patients but not in those receiving everolimus. Given the failure to replicate the development set findings in our validation cohort, further re-validation within the RCC population is warranted. However, these results enhance our understanding of the genetic predisposition to trAEs and provide a significant step toward safer and more effective cancer treatment strategies. This study was registered on the University Hospital Medical Information Network (UMIN) in Japan on August 20, 2019 (protocol ID: UMIN000037739).</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"116 12","pages":"3300-3308"},"PeriodicalIF":4.3,"publicationDate":"2025-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cas.70204","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145139142","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}
Radionuclides such as Rhenium-188 (Re188) hold promise for treating metastatic cancers due to their cytotoxic effects and potential to stimulate systemic anti-tumor immunity. However, mononuclear phagocyte system-mediated clearance of liposome encapsulated Re188 (Lipo-Re188) limits its tumor delivery. This study aimed to enhance the therapeutic effect of Lipo-Re188 against lung metastases through macrophage depletion and immune checkpoint blockade. A lung metastatic colon cancer model was established via intravenous injection of CT26-luciferase cells and then treated with Lipo-Re188 (11.1 MBq, 30% of MTD), liposomal clodronate (Lipo-clod) for macrophage depletion, and/or anti-PD-L1 antibody. Tumor progression was monitored by bioluminescence imaging, and radionuclide biodistribution was assessed at 1, 24, and 48 h post-injection. Flow cytometry was used to assess immune cell populations in the spleen and tumor microenvironment (TME). Cytokine levels were measured using a bead-based multiplex assay and analyzed by flow cytometry. Macrophage depletion significantly enhanced tumor accumulation of Lipo-Re188 while reducing hepatic uptake and prolonging survival. The combination of Lipo-clod and Lipo-Re188 promoted B cells, restored functional T cells, and suppressed MDSC in both spleen and TME. Notably, IL-1α and GM-CSF levels were significantly elevated in the combination group. Triple therapy with Lipo-clod, Lipo-Re188, and anti-PD-L1 provided the greatest survival benefit, highest intratumoral B cell accumulation, and lowest interstitial macrophage levels, with no significant biological toxicity. Our study reveals that triple therapy overcomes immunosuppressive feedback and promotes a tumor-suppressive microenvironment. These findings support a rational combination strategy integrating radiopharmaceutical therapy with immune modulation for metastatic cancer treatment.
{"title":"Liposomal 188Rhenium Plus Macrophage Depletion Enhances Anti-PD-L1 Efficacy and B Cell Infiltration Against Lung Metastatic Cancer","authors":"Shin-Yi Liu, Liang-Ting Lin, Chih-Hsien Chang, Yu-Jen Chen","doi":"10.1111/cas.70206","DOIUrl":"10.1111/cas.70206","url":null,"abstract":"<p>Radionuclides such as Rhenium-188 (Re188) hold promise for treating metastatic cancers due to their cytotoxic effects and potential to stimulate systemic anti-tumor immunity. However, mononuclear phagocyte system-mediated clearance of liposome encapsulated Re188 (Lipo-Re188) limits its tumor delivery. This study aimed to enhance the therapeutic effect of Lipo-Re188 against lung metastases through macrophage depletion and immune checkpoint blockade. A lung metastatic colon cancer model was established via intravenous injection of CT26-luciferase cells and then treated with Lipo-Re188 (11.1 MBq, 30% of MTD), liposomal clodronate (Lipo-clod) for macrophage depletion, and/or anti-PD-L1 antibody. Tumor progression was monitored by bioluminescence imaging, and radionuclide biodistribution was assessed at 1, 24, and 48 h post-injection. Flow cytometry was used to assess immune cell populations in the spleen and tumor microenvironment (TME). Cytokine levels were measured using a bead-based multiplex assay and analyzed by flow cytometry. Macrophage depletion significantly enhanced tumor accumulation of Lipo-Re188 while reducing hepatic uptake and prolonging survival. The combination of Lipo-clod and Lipo-Re188 promoted B cells, restored functional T cells, and suppressed MDSC in both spleen and TME. Notably, IL-1α and GM-CSF levels were significantly elevated in the combination group. Triple therapy with Lipo-clod, Lipo-Re188, and anti-PD-L1 provided the greatest survival benefit, highest intratumoral B cell accumulation, and lowest interstitial macrophage levels, with no significant biological toxicity. Our study reveals that triple therapy overcomes immunosuppressive feedback and promotes a tumor-suppressive microenvironment. These findings support a rational combination strategy integrating radiopharmaceutical therapy with immune modulation for metastatic cancer treatment.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"116 12","pages":"3442-3458"},"PeriodicalIF":4.3,"publicationDate":"2025-09-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cas.70206","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145114985","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}
Teng Wei, Wen Cao, Qiangnu Zhang, E. Wu, Qineng Li, Lili Ren
T-cell receptor (TCR) bispecific proteins represent a pioneering therapeutic modality that harnesses the diverse target recognition capabilities inherent to TCRs while preserving the beneficial characteristics associated with protein therapeutics. However, investigations into TCR bispecific proteins, particularly within the context of lung diseases, remain limited. This study aims to elucidate their potential for the treatment of lung cancer. We employed a humanized murine model to evaluate the efficacy of TCR bispecific proteins in eradicating lung tumors in vivo. Cytotoxic activity was assessed against various lung cancer cell lines, and statistical tests were used to analyze the data. To address concerns regarding toxic side effects from systemic administration, mesenchymal stem cells (MSCs) were explored as vehicles for the targeted delivery of TCR bispecific proteins. Our findings demonstrate that TCR bispecific proteins exhibit substantial cytotoxic activity against a variety of lung cancer cell lines. MSCs, with optimal pulmonary targeting properties, were shown to persist within the lungs for over 7 days. By employing MSCs to locally secrete TCR bispecific proteins, we achieved therapeutic effects comparable to systemic administration without manifestations of immune overactivation in murine subjects. Additionally, we evaluated various cytokine combinations and discovered that the combination of IL-7, IL-21, and TCR bispecific proteins significantly augmented their capacity to eliminate antigen-negative cells in a heterogeneous tumor model. Collectively, our findings suggest that the combinatorial therapy of TCR bispecific proteins and MSCs holds considerable promise for clinical application in the treatment of lung cancer, potentially enhancing therapeutic efficacy while minimizing adverse effects.
{"title":"Pulmonary Delivery of TCR Bispecific Proteins via Mesenchymal Stem Cells Facilitates Efficient Clearance of Lung Cancers","authors":"Teng Wei, Wen Cao, Qiangnu Zhang, E. Wu, Qineng Li, Lili Ren","doi":"10.1111/cas.70192","DOIUrl":"10.1111/cas.70192","url":null,"abstract":"<p>T-cell receptor (TCR) bispecific proteins represent a pioneering therapeutic modality that harnesses the diverse target recognition capabilities inherent to TCRs while preserving the beneficial characteristics associated with protein therapeutics. However, investigations into TCR bispecific proteins, particularly within the context of lung diseases, remain limited. This study aims to elucidate their potential for the treatment of lung cancer. We employed a humanized murine model to evaluate the efficacy of TCR bispecific proteins in eradicating lung tumors in vivo. Cytotoxic activity was assessed against various lung cancer cell lines, and statistical tests were used to analyze the data. To address concerns regarding toxic side effects from systemic administration, mesenchymal stem cells (MSCs) were explored as vehicles for the targeted delivery of TCR bispecific proteins. Our findings demonstrate that TCR bispecific proteins exhibit substantial cytotoxic activity against a variety of lung cancer cell lines. MSCs, with optimal pulmonary targeting properties, were shown to persist within the lungs for over 7 days. By employing MSCs to locally secrete TCR bispecific proteins, we achieved therapeutic effects comparable to systemic administration without manifestations of immune overactivation in murine subjects. Additionally, we evaluated various cytokine combinations and discovered that the combination of IL-7, IL-21, and TCR bispecific proteins significantly augmented their capacity to eliminate antigen-negative cells in a heterogeneous tumor model. Collectively, our findings suggest that the combinatorial therapy of TCR bispecific proteins and MSCs holds considerable promise for clinical application in the treatment of lung cancer, potentially enhancing therapeutic efficacy while minimizing adverse effects.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"116 11","pages":"3185-3195"},"PeriodicalIF":4.3,"publicationDate":"2025-09-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cas.70192","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145114950","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}
At the first interim analysis of the global, randomized, phase 3, double-blind ADRIATIC trial in patients with limited-stage small-cell lung cancer (LS-SCLC) not progressing after concurrent chemoradiotherapy (cCRT), consolidation durvalumab significantly improved overall survival (OS; hazard ratio [HR] 0.73) and progression-free survival (PFS) by blinded independent central review (BICR; HR 0.76) versus placebo (dual primary endpoints). We report an exploratory analysis in patients enrolled in Japan. Patients received durvalumab 1500 mg (N = 264), durvalumab+tremelimumab 75 mg (4 doses, N = 200; arm remained blinded), or placebo (N = 266) every 4 weeks for ≤ 24 months. Prior cCRT ± prophylactic cranial irradiation (PCI) was per local standards of care. In the Japan subgroup, 19 and 31 patients received durvalumab and placebo, respectively; prior cCRT comprised cisplatin-etoposide/carboplatin-etoposide in 94.7/5.3% and 87.1/12.9% and once-daily/twice-daily radiotherapy in 10.5/89.5% and 0/100%; 52.6% and 58.1% received PCI. Median OS was not reached versus 44.9 months (3-year OS 67.4% versus 58.1%; HR 0.67, 95% CI 0.24–1.62). Median PFS by BICR was 44.2 versus 29.4 months (24-month PFS 59.6% vs. 58.6%; HR 1.05, 95% CI 0.44–2.36); median PFS by investigator assessment (sensitivity analysis) was 44.2 versus 19.7 months (24-month PFS 65.6% vs. 47.0%; HR 0.68, 95% CI 0.28–1.51). With durvalumab and placebo, 21.1% and 19.4% had maximum grade 3–4 adverse events (AEs), 21.1% and 9.7% had AEs leading to treatment discontinuation, and 52.6% and 45.2% had pneumonitis/radiation pneumonitis (grade 3–4: 0% and 6.5%). In conclusion, consolidation durvalumab demonstrated a favorable risk/benefit profile in Japanese patients with LS-SCLC post cCRT.
在一项针对同步放化疗(cCRT)后未进展的有限期小细胞肺癌(LS-SCLC)患者的全球随机3期双盲ADRIATIC试验的首次中期分析中,通过盲法独立中心评价(BICR; HR 0.76),巩固durvalumab与安慰剂(双主要终点)相比显著提高了总生存率(OS;风险比[HR] 0.73)和无进展生存率(PFS)。我们报告了一项在日本登记的患者的探索性分析。患者每4周接受durvalumab 1500 mg (N = 264)、durvalumab+tremelimumab 75 mg(4次剂量,N = 200;对照组保持盲法)或安慰剂(N = 266)治疗,疗程≤24个月。既往cCRT±预防性颅脑照射(PCI)按当地护理标准进行。在日本亚组中,分别有19名和31名患者接受了durvalumab和安慰剂;既往cCRT包括顺铂-依托泊苷/卡铂-依托泊苷的比例分别为94.7/5.3%和87.1/12.9%,每日一次/每日两次放疗的比例分别为10.5/89.5%和0/100%;分别为52.6%和58.1%。中位OS未达到,vs . 44.9个月(3年OS 67.4% vs . 58.1%; HR 0.67, 95% CI 0.24-1.62)。BICR的中位PFS为44.2个月对29.4个月(24个月PFS为59.6%对58.6%;HR 1.05, 95% CI 0.44-2.36);研究者评估(敏感性分析)的中位PFS为44.2个月对19.7个月(24个月PFS为65.6%对47.0%;HR 0.68, 95% CI 0.28-1.51)。使用durvalumab和安慰剂,21.1%和19.4%的患者出现最大3-4级不良事件(ae), 21.1%和9.7%的患者出现导致停药的ae, 52.6%和45.2%的患者出现肺炎/放射性肺炎(3-4级:0%和6.5%)。总之,巩固durvalumab在cCRT后的日本LS-SCLC患者中显示出有利的风险/获益概况。试验注册:ClinicalTrials.gov标识符:NCT03703297。
{"title":"Durvalumab Post Concurrent Chemoradiotherapy in Japanese Patients With Limited-Stage Small-Cell Lung Cancer in the Phase 3 ADRIATIC Trial","authors":"Yoshitaka Zenke, Yoshimasa Shiraishi, Yasushi Goto, Koichi Azuma, Kyoichi Okishio, Hirokazu Ogino, Yoshitsugu Horio, Satoshi Oizumi, Manabu Hayama, Masahiro Nii, Masaya Harada, Helen Mann, Yuka S. Olivo, Haiyi Jiang, Suresh Senan","doi":"10.1111/cas.70188","DOIUrl":"10.1111/cas.70188","url":null,"abstract":"<p>At the first interim analysis of the global, randomized, phase 3, double-blind ADRIATIC trial in patients with limited-stage small-cell lung cancer (LS-SCLC) not progressing after concurrent chemoradiotherapy (cCRT), consolidation durvalumab significantly improved overall survival (OS; hazard ratio [HR] 0.73) and progression-free survival (PFS) by blinded independent central review (BICR; HR 0.76) versus placebo (dual primary endpoints). We report an exploratory analysis in patients enrolled in Japan. Patients received durvalumab 1500 mg (<i>N</i> = 264), durvalumab+tremelimumab 75 mg (4 doses, <i>N</i> = 200; arm remained blinded), or placebo (<i>N</i> = 266) every 4 weeks for ≤ 24 months. Prior cCRT ± prophylactic cranial irradiation (PCI) was per local standards of care. In the Japan subgroup, 19 and 31 patients received durvalumab and placebo, respectively; prior cCRT comprised cisplatin-etoposide/carboplatin-etoposide in 94.7/5.3% and 87.1/12.9% and once-daily/twice-daily radiotherapy in 10.5/89.5% and 0/100%; 52.6% and 58.1% received PCI. Median OS was not reached versus 44.9 months (3-year OS 67.4% versus 58.1%; HR 0.67, 95% CI 0.24–1.62). Median PFS by BICR was 44.2 versus 29.4 months (24-month PFS 59.6% vs. 58.6%; HR 1.05, 95% CI 0.44–2.36); median PFS by investigator assessment (sensitivity analysis) was 44.2 versus 19.7 months (24-month PFS 65.6% vs. 47.0%; HR 0.68, 95% CI 0.28–1.51). With durvalumab and placebo, 21.1% and 19.4% had maximum grade 3–4 adverse events (AEs), 21.1% and 9.7% had AEs leading to treatment discontinuation, and 52.6% and 45.2% had pneumonitis/radiation pneumonitis (grade 3–4: 0% and 6.5%). In conclusion, consolidation durvalumab demonstrated a favorable risk/benefit profile in Japanese patients with LS-SCLC post cCRT.</p><p><b>Trial Registration:</b> ClinicalTrials.gov identifier: NCT03703297.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"116 11","pages":"3171-3184"},"PeriodicalIF":4.3,"publicationDate":"2025-09-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cas.70188","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145114901","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}
Choriocarcinoma is a rare gynecologic malignancy. MicroRNAs, which are noncoding RNAs approximately 22 nucleotides in length, are known to regulate gene expression and play important roles in various cancers; however, their functions in choriocarcinoma remain largely unknown. This study aimed to identify disease-specific microRNAs involved in choriocarcinoma development. Eleven cases of choriocarcinoma and five cases of complete hydatidiform mole treated at our institution were analyzed. Total RNA was extracted from trophoblast cells in formalin-fixed, paraffin-embedded specimens using laser capture microdissection, and microRNA sequencing was performed. The analysis revealed that 87 microRNAs were significantly upregulated, whereas 28 were downregulated in choriocarcinoma compared to complete hydatidiform mole. Notably, 13 of the 28 downregulated microRNAs belonged to the chromosome 19 microRNA cluster. In vitro experiments demonstrated that overexpression of miR-517a-3p, a representative member of this cluster, significantly suppressed cell proliferation, migration, and invasion in JEG-3 and BeWo cell lines. Further transcriptome sequencing and computational analysis identified SRSF1 as a target gene of miR-517a-3p, which was validated by dual-luciferase reporter assays. Knockdown of SRSF1 also led to significant reductions in proliferation, migration, and invasion, supporting its functional relevance. Immunohistochemical analysis confirmed that SRSF1 protein was highly expressed in choriocarcinoma tissues compared to complete hydatidiform mole. These findings indicate that downregulation of the chromosome 19 microRNA cluster is a characteristic feature of choriocarcinoma and that miR-517a-3p functions as a tumor suppressor by directly regulating SRSF1 expression.
{"title":"Downregulation of Chromosome 19 miRNA Cluster and the Tumor-Suppressive Role of miR-517a-3p in Choriocarcinoma","authors":"Yuki Nishiko, Kosuke Yoshida, Yuko Yasui, Akira Yokoi, Masami Kitagawa, Eri Inami, Masato Yoshihara, Kazumasa Mogi, Yukari Nagao, Satoshi Tamauchi, Nobuhisa Yoshikawa, Kimihiro Nishino, Eiko Yamamoto, Kaoru Niimi, Hiroaki Kajiyama","doi":"10.1111/cas.70207","DOIUrl":"10.1111/cas.70207","url":null,"abstract":"<p>Choriocarcinoma is a rare gynecologic malignancy. MicroRNAs, which are noncoding RNAs approximately 22 nucleotides in length, are known to regulate gene expression and play important roles in various cancers; however, their functions in choriocarcinoma remain largely unknown. This study aimed to identify disease-specific microRNAs involved in choriocarcinoma development. Eleven cases of choriocarcinoma and five cases of complete hydatidiform mole treated at our institution were analyzed. Total RNA was extracted from trophoblast cells in formalin-fixed, paraffin-embedded specimens using laser capture microdissection, and microRNA sequencing was performed. The analysis revealed that 87 microRNAs were significantly upregulated, whereas 28 were downregulated in choriocarcinoma compared to complete hydatidiform mole. Notably, 13 of the 28 downregulated microRNAs belonged to the chromosome 19 microRNA cluster. In vitro experiments demonstrated that overexpression of miR-517a-3p, a representative member of this cluster, significantly suppressed cell proliferation, migration, and invasion in JEG-3 and BeWo cell lines. Further transcriptome sequencing and computational analysis identified SRSF1 as a target gene of miR-517a-3p, which was validated by dual-luciferase reporter assays. Knockdown of SRSF1 also led to significant reductions in proliferation, migration, and invasion, supporting its functional relevance. Immunohistochemical analysis confirmed that SRSF1 protein was highly expressed in choriocarcinoma tissues compared to complete hydatidiform mole. These findings indicate that downregulation of the chromosome 19 microRNA cluster is a characteristic feature of choriocarcinoma and that miR-517a-3p functions as a tumor suppressor by directly regulating SRSF1 expression.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"116 12","pages":"3473-3486"},"PeriodicalIF":4.3,"publicationDate":"2025-09-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cas.70207","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145114932","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}
Hong Tao, Luyu Wang, Yi Ding, Lixian Yi, Mutian Han, Mengmeng Gu, Jian Wu
The development of cervical cancer is strongly associated with persistent high-risk HPV infection. Microbiota and metabolomics offer new perspectives. This article focuses on the role of microbial dysbiosis and metabolic reprogramming in the development of cervical cancer, revealing its synergistic regulation of the tumor immune microenvironment and treatment resistance. Machine learning and multi-omics have identified new biomarkers, while treatment strategies include microbiota modulation, metabolic targeting, and combination therapies. Despite limitations such as small sample size and unclear mechanisms, this review proposes a multi-target precision medicine framework. In the future, we should focus on multi-center and multi-omics research and optimized clinical trials.
{"title":"Microbial and Metabolic Disorders in Cervical Cancer: Structural Insights, Biomarkers, Mechanisms, and Therapeutic Strategies","authors":"Hong Tao, Luyu Wang, Yi Ding, Lixian Yi, Mutian Han, Mengmeng Gu, Jian Wu","doi":"10.1111/cas.70201","DOIUrl":"10.1111/cas.70201","url":null,"abstract":"<p>The development of cervical cancer is strongly associated with persistent high-risk HPV infection. Microbiota and metabolomics offer new perspectives. This article focuses on the role of microbial dysbiosis and metabolic reprogramming in the development of cervical cancer, revealing its synergistic regulation of the tumor immune microenvironment and treatment resistance. Machine learning and multi-omics have identified new biomarkers, while treatment strategies include microbiota modulation, metabolic targeting, and combination therapies. Despite limitations such as small sample size and unclear mechanisms, this review proposes a multi-target precision medicine framework. In the future, we should focus on multi-center and multi-omics research and optimized clinical trials.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"116 12","pages":"3228-3238"},"PeriodicalIF":4.3,"publicationDate":"2025-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cas.70201","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145087940","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}
Antibacterial quinolones are widely used to treat bacterial infections in humans. They inhibit bacterial DNA gyrase and topoisomerase IV, whose analogous enzymes are present in mammalian mitochondria. Quinolones inhibit mitochondrial topoisomerases, thereby leading to mitochondrial DNA (mtDNA) replication suppression and cancer cell death. Meanwhile, high concentrations of quinolones are required to induce cancer cell death, possibly owing to poor delivery to the mitochondria. In this study, we synthesized nalidixic acid (NA) and ciprofloxacin (CFX) conjugated with the mitochondria-targeting moiety triphenylphosphonium (TPP), NX–TPP and CFX–TPP, to enhance mitochondrial delivery and examined their anticancer efficacy. NX–TPP and CFX–TPP markedly reduced the antibacterial activity, although CFX–TPP was more active than NX–TPP. However, both NX–TPP and CFX–TPP significantly induced cell death in colon HT-29, pancreatic MIAPaCa-2, and other cancer cells but not in non-cancerous cells including normal dermal fibroblasts and human vascular endothelial cells at a comparative level. NX–TPP induced necrosis-like cell death characterized by cell membrane ballooning and rupture. Mechanistically, NX–TPP was efficiently incorporated into the mitochondria, leading to increased mitochondrial reaction oxygen species (mtROS) generation and mitophagy, and decreased mtDNA copy number and mitochondrial respiration. NX–TPP inhibited tumor growth in HT-29 and MIAPaCa-2 xenograft mouse models without any apparent adverse effects. These results suggest that mtDNA replication-targeting quinolone derivatives, termed MitoQNs, that exhibit reduced antibacterial activity, thereby decreasing antibiotic resistance induction, and enhanced anticancer efficacy, are candidate drugs for cancer therapy.
{"title":"Anticancer Effect of the Triphenylphosphonium-Conjugated Quinolone Antibiotics Targeting Mitochondrial DNA Replication","authors":"Yuming Qiao, Yuki Kida, Xiaoyi Lai, Nobuko Koshikawa, Rie Igarashi, Atsushi Takatori, Keizo Takenaga","doi":"10.1111/cas.70199","DOIUrl":"10.1111/cas.70199","url":null,"abstract":"<p>Antibacterial quinolones are widely used to treat bacterial infections in humans. They inhibit bacterial DNA gyrase and topoisomerase IV, whose analogous enzymes are present in mammalian mitochondria. Quinolones inhibit mitochondrial topoisomerases, thereby leading to mitochondrial DNA (mtDNA) replication suppression and cancer cell death. Meanwhile, high concentrations of quinolones are required to induce cancer cell death, possibly owing to poor delivery to the mitochondria. In this study, we synthesized nalidixic acid (NA) and ciprofloxacin (CFX) conjugated with the mitochondria-targeting moiety triphenylphosphonium (TPP), NX–TPP and CFX–TPP, to enhance mitochondrial delivery and examined their anticancer efficacy. NX–TPP and CFX–TPP markedly reduced the antibacterial activity, although CFX–TPP was more active than NX–TPP. However, both NX–TPP and CFX–TPP significantly induced cell death in colon HT-29, pancreatic MIAPaCa-2, and other cancer cells but not in non-cancerous cells including normal dermal fibroblasts and human vascular endothelial cells at a comparative level. NX–TPP induced necrosis-like cell death characterized by cell membrane ballooning and rupture. Mechanistically, NX–TPP was efficiently incorporated into the mitochondria, leading to increased mitochondrial reaction oxygen species (mtROS) generation and mitophagy, and decreased mtDNA copy number and mitochondrial respiration. NX–TPP inhibited tumor growth in HT-29 and MIAPaCa-2 xenograft mouse models without any apparent adverse effects. These results suggest that mtDNA replication-targeting quinolone derivatives, termed MitoQNs, that exhibit reduced antibacterial activity, thereby decreasing antibiotic resistance induction, and enhanced anticancer efficacy, are candidate drugs for cancer therapy.</p>","PeriodicalId":9580,"journal":{"name":"Cancer Science","volume":"116 12","pages":"3431-3441"},"PeriodicalIF":4.3,"publicationDate":"2025-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cas.70199","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145076324","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}