Nab-paclitaxel plus gemcitabine (AG) is the standard first-line chemotherapy for advanced or metastatic pancreatic ductal adenocarcinoma and has limited efficacy. This phase 1b/2 study aimed to evaluate SHR-1701 (an anti-PD-L1/TGF-βRII fusion protein) plus AG in this population (NCT04624217). In phase 1b part, the recommended dose of SHR-1701 was identified as 30 mg/kg every 3 weeks, when combined with AG. In phase 2 part, the primary endpoint was objective response rate (ORR). As of Mar 31, 2023, 56 patients were enrolled. Median follow-up was 10.3 months (range, 0.2-24.7). ORR was 32.1% (95% CI, 20.3-46.0). Median progressive-free survival (PFS) was 5.6 months (95% CI, 4.3-6.6), and median overall survival (OS) was 10.3 months (95% CI, 8.8-12.3). Treatment-related adverse events of grade ≥3 were reported in 27 (48.2%) patients, with the most common being decreased neutrophil count. Patients with PD-L1 TPS ≥ 1% showed a higher ORR (66.7% vs. 25.0%), as well as extended median PFS (6.3 vs. 5.3 months) and median OS (18.8 vs. 9.9 months). Additionally, reduction of CA19-9 by at least 80% during treatment and pSMAD2/3 staining intensity of 1+ at baseline were potential monitoring tools and predictive biomarkers for better clinical outcomes, respectively. Tumor-specific T-cell infiltration and pancreatic cancer tumor subtypes were associated with anti-tumor response. The interactions within tumor microenvironment were involved disease progression. Overall, first-line SHR-1701 plus AG showed promising anti-tumor activity and controllable safety in advanced or metastatic pancreatic ductal adenocarcinoma, and features of patients more likely to benefit from the combination were drawn.
nab -紫杉醇加吉西他滨(AG)是晚期或转移性胰腺导管腺癌的标准一线化疗方案,但疗效有限。这项1b/2期研究旨在评估SHR-1701(一种抗pd - l1 /TGF-βRII融合蛋白)加AG在该人群中的作用(NCT04624217)。在1b期研究中,SHR-1701的推荐剂量确定为每3周30 mg/kg,与AG联合使用。在第二阶段,主要终点是客观缓解率(ORR)。截至2023年3月31日,共有56名患者入组。中位随访时间为10.3个月(范围0.2-24.7)。ORR为32.1% (95% CI, 20.3-46.0)。中位无进展生存期(PFS)为5.6个月(95% CI, 4.3-6.6),中位总生存期(OS)为10.3个月(95% CI, 8.8-12.3)。27例(48.2%)患者报告了≥3级的治疗相关不良事件,最常见的是中性粒细胞计数减少。PD-L1 TPS≥1%的患者表现出更高的ORR (66.7% vs. 25.0%),中位PFS (6.3 vs. 5.3个月)和中位OS (18.8 vs. 9.9个月)延长。此外,治疗期间CA19-9减少至少80%,基线时pSMAD2/3染色强度为1+,分别是更好临床结果的潜在监测工具和预测性生物标志物。肿瘤特异性t细胞浸润和胰腺癌肿瘤亚型与抗肿瘤反应相关。肿瘤微环境内的相互作用与疾病进展有关。总体而言,一线SHR-1701联合AG在晚期或转移性胰腺导管腺癌中显示出良好的抗肿瘤活性和可控的安全性,并且更有可能从联合治疗中获益的患者的特征被描绘出来。
{"title":"A phase 1b/2 study of first-line anti-PD-L1/ TGF-βRII fusion protein SHR-1701 combined with nab-paclitaxel and gemcitabine for advanced pancreatic ductal adenocarcinoma.","authors":"Ran Xue,Miaoyan Wei,Jiajia Yuan,Zhihua Li,Yuhong Zhou,Zeyun Xue,Yiwen Wu,Hongxia Han,Jun Zhou,Xianjun Yu,Lin Shen","doi":"10.1038/s41392-025-02530-2","DOIUrl":"https://doi.org/10.1038/s41392-025-02530-2","url":null,"abstract":"Nab-paclitaxel plus gemcitabine (AG) is the standard first-line chemotherapy for advanced or metastatic pancreatic ductal adenocarcinoma and has limited efficacy. This phase 1b/2 study aimed to evaluate SHR-1701 (an anti-PD-L1/TGF-βRII fusion protein) plus AG in this population (NCT04624217). In phase 1b part, the recommended dose of SHR-1701 was identified as 30 mg/kg every 3 weeks, when combined with AG. In phase 2 part, the primary endpoint was objective response rate (ORR). As of Mar 31, 2023, 56 patients were enrolled. Median follow-up was 10.3 months (range, 0.2-24.7). ORR was 32.1% (95% CI, 20.3-46.0). Median progressive-free survival (PFS) was 5.6 months (95% CI, 4.3-6.6), and median overall survival (OS) was 10.3 months (95% CI, 8.8-12.3). Treatment-related adverse events of grade ≥3 were reported in 27 (48.2%) patients, with the most common being decreased neutrophil count. Patients with PD-L1 TPS ≥ 1% showed a higher ORR (66.7% vs. 25.0%), as well as extended median PFS (6.3 vs. 5.3 months) and median OS (18.8 vs. 9.9 months). Additionally, reduction of CA19-9 by at least 80% during treatment and pSMAD2/3 staining intensity of 1+ at baseline were potential monitoring tools and predictive biomarkers for better clinical outcomes, respectively. Tumor-specific T-cell infiltration and pancreatic cancer tumor subtypes were associated with anti-tumor response. The interactions within tumor microenvironment were involved disease progression. Overall, first-line SHR-1701 plus AG showed promising anti-tumor activity and controllable safety in advanced or metastatic pancreatic ductal adenocarcinoma, and features of patients more likely to benefit from the combination were drawn.","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"181 1","pages":"415"},"PeriodicalIF":39.3,"publicationDate":"2025-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145796241","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-19DOI: 10.1038/s41392-025-02505-3
Mengyao Xu,Syem K Barakzai,Raj Kumar,Irva Veillard,Eugene Kim,Amy Bregar,Eric Eisenhauer,Richard Penson,Sara Bouberhan,Jennifer Filipi,Tina Colella,Tim Bond,Caroline Clark,Lawrence H Lin,Jinpeng Ruan,Cheng Wang,Xingping Qin,Kristopher Sarosiek,Bo Rueda,Cesar Castro,David R Spriggs,Oladapo O Yeku
Bispecific T-cell Engagers (BITEs) are a novel form of immunotherapy that overcome a deficiency of immune checkpoint inhibitors (ICI) by targeting a preidentified tumor associated antigen and redirecting a polyclonal population of effector T-cells against the tumor. High grade serous ovarian cancer is a lethal disease in the recurrent setting and has not been amenable to ICI therapy. MUC16/CA125 is overexpressed in high grade serous ovarian cancer. BITEs targeting the tumor-retained portion of MUC16/CA125 have recently been described and are in early-phase clinical trials. To identify mechanisms of resistance to BITEs, we collected serum, peripheral blood mononuclear cells, and ascites samples from patients with disease progression on MUC16-directed bispecific antibodies. Analysis of these samples showed downregulation of MUC16/CA125, elevated secretion of VEGF, and epithelial-to-mesenchymal transition in tumor cells. Interestingly, hypoxia was determined to be a driver of these changes. These findings were prospectively validated in ovarian cancer cell lines with CRISPR/Cas9 knockout of MUC16/CA125 and VEGF. Peripheral blood mononuclear cells from patients with disease progression were capable of effective cytolysis ex vivo, suggesting that resistance to therapy was primarily tumor driven. Restoration of MUC16/CA125 expression did not restore cytotoxicity in the presence of increased VEGF secretion. Combination treatment with a VEGF inhibitor rescued cytotoxicity in hypoxia-conditioned ovarian cancer cell lines with preserved target antigen expression. Collectively, these data outline a link between hypoxia and the development of resistance to BITEs and posits inhibition of VEGF inhibition as a potentially important therapeutic intervention.
{"title":"Hypoxia induced VEGF secretion promotes resistance to bispecific T-cell engagers.","authors":"Mengyao Xu,Syem K Barakzai,Raj Kumar,Irva Veillard,Eugene Kim,Amy Bregar,Eric Eisenhauer,Richard Penson,Sara Bouberhan,Jennifer Filipi,Tina Colella,Tim Bond,Caroline Clark,Lawrence H Lin,Jinpeng Ruan,Cheng Wang,Xingping Qin,Kristopher Sarosiek,Bo Rueda,Cesar Castro,David R Spriggs,Oladapo O Yeku","doi":"10.1038/s41392-025-02505-3","DOIUrl":"https://doi.org/10.1038/s41392-025-02505-3","url":null,"abstract":"Bispecific T-cell Engagers (BITEs) are a novel form of immunotherapy that overcome a deficiency of immune checkpoint inhibitors (ICI) by targeting a preidentified tumor associated antigen and redirecting a polyclonal population of effector T-cells against the tumor. High grade serous ovarian cancer is a lethal disease in the recurrent setting and has not been amenable to ICI therapy. MUC16/CA125 is overexpressed in high grade serous ovarian cancer. BITEs targeting the tumor-retained portion of MUC16/CA125 have recently been described and are in early-phase clinical trials. To identify mechanisms of resistance to BITEs, we collected serum, peripheral blood mononuclear cells, and ascites samples from patients with disease progression on MUC16-directed bispecific antibodies. Analysis of these samples showed downregulation of MUC16/CA125, elevated secretion of VEGF, and epithelial-to-mesenchymal transition in tumor cells. Interestingly, hypoxia was determined to be a driver of these changes. These findings were prospectively validated in ovarian cancer cell lines with CRISPR/Cas9 knockout of MUC16/CA125 and VEGF. Peripheral blood mononuclear cells from patients with disease progression were capable of effective cytolysis ex vivo, suggesting that resistance to therapy was primarily tumor driven. Restoration of MUC16/CA125 expression did not restore cytotoxicity in the presence of increased VEGF secretion. Combination treatment with a VEGF inhibitor rescued cytotoxicity in hypoxia-conditioned ovarian cancer cell lines with preserved target antigen expression. Collectively, these data outline a link between hypoxia and the development of resistance to BITEs and posits inhibition of VEGF inhibition as a potentially important therapeutic intervention.","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"16 1","pages":"405"},"PeriodicalIF":39.3,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145777316","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
CDK2 is a principal mediator of CDK4/6 resistance. Concurrent CDK2/4/6 blockade may be effective in treating HR-positive, HER2-negative advanced breast cancer (ABC). This randomized, double-blind, parallel-controlled, phase 3 trial (ClinicalTrials.gov, NCT05375461) assessed the efficacy of culmerciclib, a CDK2/4/6 inhibitor, plus fulvestrant in ABC. Patients with HR-positive, HER2-negative, locally recurrent or metastatic breast cancer were randomized (2:1) to receive culmerciclib plus fulvestrant or matching placebo plus fulvestrant. Between March 18, 2022 and March 3, 2023, 293 pretreated patients (median age 53.0 years; pre- or perimenopausal 42.3%; bone metastasis 65.2%) were randomized to assigned treatments. At this prespecified interim analysis, culmerciclib plus fulvestrant extended the median investigator-assessed progression-free survival (PFS) significantly, the primary endpoint, as compared with placebo plus fulvestrant (16.6 months, 95% CI 13.8 to not evaluable versus 7.5 months, 95% CI 5.3 to 11.0; hazard ratio 0.36, 95% CI 0.26–0.51; stratified log rank test P < 0.001). Consistent effects were observed across diverse subgroups of patients. At a median follow-up duration of 13.8 months, overall survival was immature. The investigators-assessed objective response rate was 40.2% (95% CI, 33.3–47.5) for culmerciclib compared to 12.1% (95% CI 6.4–20.2) for placebo (stratified Mantel-Haenszel χ 2 test P < 0.001). Diarrhea (87.1%) and neutropenia (80.4%) were the most common toxicities with culmerciclib plus fulvestrant. In conclusion, this randomized clinical trial met its primary outcome. Culmerciclib plus fulvestrant is well tolerated and leads to a significant gain in PFS of pretreated HR-positive HER2-negative ABC patients.
CDK2是CDK4/6耐药的主要媒介。同时阻断CDK2/4/6可能对hr阳性、her2阴性的晚期乳腺癌(ABC)有效。这项随机、双盲、平行对照的3期试验(ClinicalTrials.gov, NCT05375461)评估了culmerciclib(一种CDK2/4/6抑制剂)联合氟维司汀治疗ABC的疗效。hr阳性、her2阴性、局部复发或转移性乳腺癌患者随机(2:1)接受卡美西尼加氟维司汀或匹配的安慰剂加氟维司汀。在2022年3月18日至2023年3月3日期间,293例预先治疗的患者(中位年龄53.0岁;绝经前或围绝经期42.3%;骨转移65.2%)被随机分配到指定的治疗方案。在这个预先指定的中期分析中,与安慰剂加氟维司汀相比,卡美昔布加氟维司汀显著延长了研究人员评估的无进展生存(PFS)中位数,这是主要终点(16.6个月,95% CI 13.8至不可评估,而7.5个月,95% CI 5.3至11.0;风险比0.36,95% CI 0.26-0.51;分层log rank检验P <; 0.001)。在不同亚组患者中观察到一致的效果。中位随访时间为13.8个月,总体生存期不成熟。研究者评估的客观缓解率为40.2% (95% CI, 33.3-47.5),而安慰剂为12.1% (95% CI 6.4-20.2)(分层Mantel-Haenszel χ 2检验P <; 0.001)。腹泻(87.1%)和中性粒细胞减少(80.4%)是库美西布联合氟维司汀最常见的毒性。总之,这项随机临床试验达到了其主要结局。Culmerciclib + fulvestrant具有良好的耐受性,可显著提高经预处理的hr阳性her2阴性ABC患者的PFS。
{"title":"Novel CDK2/4/6 inhibitor culmerciclib (TQB3616) plus fulvestrant in previously treated, HR-positive, HER2-negative advanced breast cancer: a randomized, double-blind, phase 3 trial","authors":"Yongmei Yin, Qingyuan Zhang, Tao Sun, Chunfang Hao, Zhihong Wang, Jin Yang, Yongsheng Wang, Yanxia Shi, Jing Sun, Quchang Ouyang, Haichuan Su, Jinsheng Wu, Lu Gan, Meng Han, Liming Gao, Xiaojia Wang, Bing Zhao, Hui Li, Jiuda Zhao, Hongwei Yang, Fangling Ning, Fuguo Tian, Juliang Zhang, Hongmei Sun, Zhaofeng Niu, Hong Zong, Aimin Zang, Xinshuai Wang, Xinyu Qian, Shikai Wu, Jianyun Nie, Lijia He, Ying Cheng, Yanrong Hao, Yi Zhai, Huiping Li, Jingfen Wang, Shihong Wei, Man Li, Yunjiang Liu, Hongqiang Guo, Qun Hu, Lina Liu, Xinghua Han, Ruizhen Luo, Mingli Ni, Xianjun Tang, Zhenhua Zhai, Meiqian Ding, Haibo Wang, Peng Shen, Xian Wang, Lian Liu, Wenyan Chen, Gang Liu, Zhengwen Cai, Zefei Jiang","doi":"10.1038/s41392-025-02475-6","DOIUrl":"https://doi.org/10.1038/s41392-025-02475-6","url":null,"abstract":"CDK2 is a principal mediator of CDK4/6 resistance. Concurrent CDK2/4/6 blockade may be effective in treating HR-positive, HER2-negative advanced breast cancer (ABC). This randomized, double-blind, parallel-controlled, phase 3 trial (ClinicalTrials.gov, NCT05375461) assessed the efficacy of culmerciclib, a CDK2/4/6 inhibitor, plus fulvestrant in ABC. Patients with HR-positive, HER2-negative, locally recurrent or metastatic breast cancer were randomized (2:1) to receive culmerciclib plus fulvestrant or matching placebo plus fulvestrant. Between March 18, 2022 and March 3, 2023, 293 pretreated patients (median age 53.0 years; pre- or perimenopausal 42.3%; bone metastasis 65.2%) were randomized to assigned treatments. At this prespecified interim analysis, culmerciclib plus fulvestrant extended the median investigator-assessed progression-free survival (PFS) significantly, the primary endpoint, as compared with placebo plus fulvestrant (16.6 months, 95% CI 13.8 to not evaluable <jats:italic>versus</jats:italic> 7.5 months, 95% CI 5.3 to 11.0; hazard ratio 0.36, 95% CI 0.26–0.51; stratified log rank test <jats:italic>P</jats:italic> < 0.001). Consistent effects were observed across diverse subgroups of patients. At a median follow-up duration of 13.8 months, overall survival was immature. The investigators-assessed objective response rate was 40.2% (95% CI, 33.3–47.5) for culmerciclib compared to 12.1% (95% CI 6.4–20.2) for placebo (stratified Mantel-Haenszel χ <jats:sup>2</jats:sup> test <jats:italic>P</jats:italic> < 0.001). Diarrhea (87.1%) and neutropenia (80.4%) were the most common toxicities with culmerciclib plus fulvestrant. In conclusion, this randomized clinical trial met its primary outcome. Culmerciclib plus fulvestrant is well tolerated and leads to a significant gain in PFS of pretreated HR-positive HER2-negative ABC patients.","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"167 1","pages":""},"PeriodicalIF":39.3,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145770703","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-17DOI: 10.1038/s41392-025-02527-x
Minjeong Jang, Hyung Jin Choi, Hae-June Lee, Hong Nam Kim
Hypoglycemia is a frequent and potentially severe complication that can result in significant brain injury in individuals with diabetes treated with insulin or other hypoglycemic agents and in those undergoing prolonged fasting. Despite its clinical importance, the molecular mechanisms through which hypoglycemia induces neurodegeneration remain poorly defined. We therefore investigated the molecular and cellular basis of hypoglycemia-induced brain damage using human neuron and glial cell cultures in vitro and hypoglycemic mouse models in vivo. We found that starvation-induced hypoglycemia triggers hallmark neurodegenerative features, such as astrocyte activation and microglial reactivity, that closely resemble those found in the brains of hypoglycemic mouse models. Neurons notably activate an adaptive survival response mediated by serum response factor (SRF) and myocardin-related transcription factor-A (MRTF-A), which drives a metabolic reprogramming process. This shift enables neurons to use extracellular matrix components as alternative energy sources under glucose deprivation. However, this compensatory mechanism results in the excessive accumulation of urea cycle byproducts, which subsequently exacerbates neuronal damage and promotes glial activation. Glucose refeeding remarkably reversed these neurodegenerative features by deactivating SRF/MRTF-A signaling in both in vitro and in vivo. Collectively, our results revealed a neuron-intrinsic mechanism linking glucose deprivation to reversible neurodegeneration via SRF/MRTF-A, offering potential targets for preventing hypoglycemia-associated brain damage.
低血糖是一种常见的、潜在的严重并发症,在接受胰岛素或其他降糖药治疗的糖尿病患者和长期禁食的糖尿病患者中可导致严重的脑损伤。尽管它的临床重要性,通过低血糖诱导神经变性的分子机制仍然不清楚。因此,我们利用体外培养的人类神经元和神经胶质细胞和体内低血糖小鼠模型来研究低血糖诱导脑损伤的分子和细胞基础。我们发现,饥饿引起的低血糖会触发标志性的神经退行性特征,如星形胶质细胞激活和小胶质细胞反应,这些特征与低血糖小鼠模型中的大脑非常相似。神经元激活由血清反应因子(SRF)和心肌素相关转录因子- a (MRTF-A)介导的适应性生存反应,从而驱动代谢重编程过程。这种转变使神经元能够利用细胞外基质成分作为葡萄糖剥夺下的替代能量来源。然而,这种代偿机制导致尿素循环副产物的过度积累,从而加剧神经元损伤并促进胶质细胞激活。在体外和体内,葡萄糖再喂养通过使SRF/MRTF-A信号失活,显著逆转了这些神经退行性特征。总的来说,我们的研究结果揭示了通过SRF/MRTF-A将葡萄糖剥夺与可逆性神经变性联系起来的神经元内在机制,为预防低血糖相关的脑损伤提供了潜在的靶点。
{"title":"Hypoglycemia induces brain metabolic reprogramming and neurodegeneration via serum response factor and myocardin-related transcription factor-A","authors":"Minjeong Jang, Hyung Jin Choi, Hae-June Lee, Hong Nam Kim","doi":"10.1038/s41392-025-02527-x","DOIUrl":"https://doi.org/10.1038/s41392-025-02527-x","url":null,"abstract":"Hypoglycemia is a frequent and potentially severe complication that can result in significant brain injury in individuals with diabetes treated with insulin or other hypoglycemic agents and in those undergoing prolonged fasting. Despite its clinical importance, the molecular mechanisms through which hypoglycemia induces neurodegeneration remain poorly defined. We therefore investigated the molecular and cellular basis of hypoglycemia-induced brain damage using human neuron and glial cell cultures in vitro and hypoglycemic mouse models in vivo. We found that starvation-induced hypoglycemia triggers hallmark neurodegenerative features, such as astrocyte activation and microglial reactivity, that closely resemble those found in the brains of hypoglycemic mouse models. Neurons notably activate an adaptive survival response mediated by serum response factor (SRF) and myocardin-related transcription factor-A (MRTF-A), which drives a metabolic reprogramming process. This shift enables neurons to use extracellular matrix components as alternative energy sources under glucose deprivation. However, this compensatory mechanism results in the excessive accumulation of urea cycle byproducts, which subsequently exacerbates neuronal damage and promotes glial activation. Glucose refeeding remarkably reversed these neurodegenerative features by deactivating SRF/MRTF-A signaling in both in vitro and in vivo. Collectively, our results revealed a neuron-intrinsic mechanism linking glucose deprivation to reversible neurodegeneration <jats:italic>via</jats:italic> SRF/MRTF-A, offering potential targets for preventing hypoglycemia-associated brain damage.","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"1 1","pages":""},"PeriodicalIF":39.3,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145770718","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-17DOI: 10.1038/s41392-025-02528-w
Liu-Zheng Wu, Ya-Ying Huang, Hai-Jing Hu, Wen-Bin Hong, Han Yan, Yuan-Li Ai, Xiang-Yu Mi, De-Yi Feng, Jian-Yi Guo, Yang Ding, Zai-Jun Liu, Bo Zhou, Li Xiao, Tianwei Lin, Fu-Nan Li, Xue-Qin Chen, Hang-Zi Chen, Qiao Wu
Pyroptosis plays a crucial role in physiological and pathological processes. As melanoma cells are resistant to apoptosis but express gasdermin proteins, it is appealing to counter melanoma with the induction of gasdermin-executed pyroptosis. GSDMC, initially cloned from metastatic melanoma cells, has been demonstrated as a potential executioner of pyroptosis. However, no lead compounds that trigger GSDMC-mediated pyroptosis have been reported, which limits the in-depth investigation of GSDMC functions. Here, we discovered a chemical compound, dodecyl 1H-benzo[d]imidazole-5-carboxylate (DdBIC), that targeted the nuclear receptor Nur77 to induce pyroptosis through cleaving GSDMC by granzyme B in melanoma cells. Upon DdBIC binding, Nur77 was translocated to the mitochondria to activate the hemoprotein SDHA to overconsume succinyl-CoA, subsequently disrupting the homeostasis of heme in the SDH complex and resulting in electron leakage to induce mito-ROS production. This mito-ROS signal was sensed by the mitochondrial protease OMA1 via oxidation, which led to downstream OPA1 cleavage and subsequent released into the cytoplasm. Cytosolic OPA1 activated PERK to induce the integrated stress response (ISR), which further activated granzyme B to cleave GSDMC, culminating in the induction of pyroptosis. Together, this study elucidates a signal cascade from Nur77-impaired homeostasis of heme metabolism to PERK-mediated ISR activation, and reveals a novel paradigm, by which granzyme B, rather than caspases, cleaves GSDMC for pyroptotic induction and provides a new strategy for the therapeutic treatment of melanoma by lead compound DdBIC.
{"title":"Disruption of heme homeostasis by nuclear receptor Nur77 induces pyroptosis through granzyme B-dependent GSDMC cleavage","authors":"Liu-Zheng Wu, Ya-Ying Huang, Hai-Jing Hu, Wen-Bin Hong, Han Yan, Yuan-Li Ai, Xiang-Yu Mi, De-Yi Feng, Jian-Yi Guo, Yang Ding, Zai-Jun Liu, Bo Zhou, Li Xiao, Tianwei Lin, Fu-Nan Li, Xue-Qin Chen, Hang-Zi Chen, Qiao Wu","doi":"10.1038/s41392-025-02528-w","DOIUrl":"https://doi.org/10.1038/s41392-025-02528-w","url":null,"abstract":"Pyroptosis plays a crucial role in physiological and pathological processes. As melanoma cells are resistant to apoptosis but express gasdermin proteins, it is appealing to counter melanoma with the induction of gasdermin-executed pyroptosis. GSDMC, initially cloned from metastatic melanoma cells, has been demonstrated as a potential executioner of pyroptosis. However, no lead compounds that trigger GSDMC-mediated pyroptosis have been reported, which limits the in-depth investigation of GSDMC functions. Here, we discovered a chemical compound, dodecyl 1H-benzo[d]imidazole-5-carboxylate (DdBIC), that targeted the nuclear receptor Nur77 to induce pyroptosis through cleaving GSDMC by granzyme B in melanoma cells. Upon DdBIC binding, Nur77 was translocated to the mitochondria to activate the hemoprotein SDHA to overconsume succinyl-CoA, subsequently disrupting the homeostasis of heme in the SDH complex and resulting in electron leakage to induce mito-ROS production. This mito-ROS signal was sensed by the mitochondrial protease OMA1 via oxidation, which led to downstream OPA1 cleavage and subsequent released into the cytoplasm. Cytosolic OPA1 activated PERK to induce the integrated stress response (ISR), which further activated granzyme B to cleave GSDMC, culminating in the induction of pyroptosis. Together, this study elucidates a signal cascade from Nur77-impaired homeostasis of heme metabolism to PERK-mediated ISR activation, and reveals a novel paradigm, by which granzyme B, rather than caspases, cleaves GSDMC for pyroptotic induction and provides a new strategy for the therapeutic treatment of melanoma by lead compound DdBIC.","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"22 1","pages":""},"PeriodicalIF":39.3,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145770705","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Wnt and its crosstalk signaling pathways are involved in the modulating ischemia‒reperfusion (I/R) injury. However, whether Wnt2 is a novel therapeutic agent for I/R injury is largely unknown. Here, we show that the downregulation of serum Wnt2 levels in acute myocardial infarction (AMI) patients following reperfusion therapy, and Wnt2 levels are inversely correlated with the levels of myocardial injury markers (cTnT and CK-MB). Therapeutic administration of recombinant Wnt2 protein (rbWnt2) alleviates cardiac I/R injury and improves cardiac function by suppressing ROS levels and cardiomyocyte death in mice. Further analysis revealed that rbWnt2 downregulated Nap1L1 to reactivate the transcription of antioxidant genes (SOD, GPX, and UCP3) to reduce ROS levels and subsequently inhibit cardiomyocyte apoptosis and ferroptosis during the I/R process. Cardiac-specific Nap1L1 knockdown attenuated I/R injury, whereas overexpression of Nap1L1 partly abolished the cardiac protection mediated by rbWnt2 administration in the I/R model. Mechanistically, Wnt2 promoted Nap1L1 ubiquitination and degradation to restore ROS scavenging systems via Lrp6-mediated recruitment of the E3 ligase Trim11 in I/R hearts. Nap1L1 suppression plays a critical role in mediating the cardioprotective effects of rbWnt2. These findings establish Wnt2 as a therapeutic agent that targets compartmentalized oxidative damage, suggesting a novel strategy to mitigate I/R injury through the Lrp6/Trim11/Nap1L1 axis.
{"title":"Nucleosome assembly protein-like 1 degradation-dependent novel cardioprotection mechanism of Wnt2 against ischemia‒reperfusion injury.","authors":"Ying Wang,Liming Chen,Jinyi Lin,Xi Liu,Kejia Jin,Chenxing Huang,Hao Wang,Jianguo Jia,Jian Wu,Zhiwen Ding,Pan Gao,Junbo Ge,Hui Gong,Yunzeng Zou","doi":"10.1038/s41392-025-02503-5","DOIUrl":"https://doi.org/10.1038/s41392-025-02503-5","url":null,"abstract":"Wnt and its crosstalk signaling pathways are involved in the modulating ischemia‒reperfusion (I/R) injury. However, whether Wnt2 is a novel therapeutic agent for I/R injury is largely unknown. Here, we show that the downregulation of serum Wnt2 levels in acute myocardial infarction (AMI) patients following reperfusion therapy, and Wnt2 levels are inversely correlated with the levels of myocardial injury markers (cTnT and CK-MB). Therapeutic administration of recombinant Wnt2 protein (rbWnt2) alleviates cardiac I/R injury and improves cardiac function by suppressing ROS levels and cardiomyocyte death in mice. Further analysis revealed that rbWnt2 downregulated Nap1L1 to reactivate the transcription of antioxidant genes (SOD, GPX, and UCP3) to reduce ROS levels and subsequently inhibit cardiomyocyte apoptosis and ferroptosis during the I/R process. Cardiac-specific Nap1L1 knockdown attenuated I/R injury, whereas overexpression of Nap1L1 partly abolished the cardiac protection mediated by rbWnt2 administration in the I/R model. Mechanistically, Wnt2 promoted Nap1L1 ubiquitination and degradation to restore ROS scavenging systems via Lrp6-mediated recruitment of the E3 ligase Trim11 in I/R hearts. Nap1L1 suppression plays a critical role in mediating the cardioprotective effects of rbWnt2. These findings establish Wnt2 as a therapeutic agent that targets compartmentalized oxidative damage, suggesting a novel strategy to mitigate I/R injury through the Lrp6/Trim11/Nap1L1 axis.","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"13 1","pages":"403"},"PeriodicalIF":39.3,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145759939","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cellular senescence is deeply involved in physiological homeostasis, development, tissue repair, aging, and diseases. Senescent cells (SnCs) accumulate in aged tissues and exert deleterious effects by secreting proinflammatory molecules that contribute to chronic inflammation and aging-related diseases. We revealed that an aberrant interaction between glycolytic PGAM1 and Chk1 kinase is augmented in SnCs associated with increased glycolysis, whose byproduct, lactate, promotes this binding in a noncell autonomous manner. The pseudo-Warburg effect of SnCs with enhanced PPP (pentose phosphate pathway) activity is maintained by HIF-2α phosphorylation by Chk1 and subsequent upregulation of glycolytic enzymes, creating a vicious cycle reprogramming the glycolytic pathway in SnCs. HIF-2α also activates FoxM1 expression, which transcriptionally suppresses proapoptotic profiles, including BIM, and upregulates DNA repair machineries in SnCs. FoxM1 thus supports the genomic integrity and survival capacity of SnCs during their glycolytic changes. Chemical abrogation of PGAM1-Chk1 binding reverts these phenotypes and eliminates SnCs through senolysis. Inhibition of the PGAM1-Chk1 interaction improves physiological parameters during aging and inhibits lung fibrosis in mouse models. Our study highlights a novel pathway contributing to the metabolic reprogramming of SnCs and how the use of a new senolytic molecule that targets the PGAM-Chk1 interaction creates a specific vulnerability of those cells to potentially fight age-related diseases.
{"title":"Abrogation of aberrant glycolytic interactions eliminates senescent cells and alleviates aging-related dysfunctions.","authors":"Takumi Mikawa,Masahiro Kameda,Sumiko Ikari,Eri Shibata,Shuyu Liu,Sawa Miyagawa,Koh Ono,Tomiko Ito,Akihiko Yoshizawa,Masataka Sugimoto,Shuichi Shibuya,Takahiko Shimizu,Julio Almunia,Noboru Ogiso,Gwladys Revêchon,Alberta Palazzo,David Bernard,Hiroaki Kanda,Tomoyoshi Soga,Keiyo Takubo,Shin Morioka,Junko Sasaki,Takehiko Sasaki,Akihiro Itamoto,Takayuki Fujii,Hiroshi Seno,Nobuya Inagaki,Hiroshi Kondoh","doi":"10.1038/s41392-025-02502-6","DOIUrl":"https://doi.org/10.1038/s41392-025-02502-6","url":null,"abstract":"Cellular senescence is deeply involved in physiological homeostasis, development, tissue repair, aging, and diseases. Senescent cells (SnCs) accumulate in aged tissues and exert deleterious effects by secreting proinflammatory molecules that contribute to chronic inflammation and aging-related diseases. We revealed that an aberrant interaction between glycolytic PGAM1 and Chk1 kinase is augmented in SnCs associated with increased glycolysis, whose byproduct, lactate, promotes this binding in a noncell autonomous manner. The pseudo-Warburg effect of SnCs with enhanced PPP (pentose phosphate pathway) activity is maintained by HIF-2α phosphorylation by Chk1 and subsequent upregulation of glycolytic enzymes, creating a vicious cycle reprogramming the glycolytic pathway in SnCs. HIF-2α also activates FoxM1 expression, which transcriptionally suppresses proapoptotic profiles, including BIM, and upregulates DNA repair machineries in SnCs. FoxM1 thus supports the genomic integrity and survival capacity of SnCs during their glycolytic changes. Chemical abrogation of PGAM1-Chk1 binding reverts these phenotypes and eliminates SnCs through senolysis. Inhibition of the PGAM1-Chk1 interaction improves physiological parameters during aging and inhibits lung fibrosis in mouse models. Our study highlights a novel pathway contributing to the metabolic reprogramming of SnCs and how the use of a new senolytic molecule that targets the PGAM-Chk1 interaction creates a specific vulnerability of those cells to potentially fight age-related diseases.","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"20 1","pages":"402"},"PeriodicalIF":39.3,"publicationDate":"2025-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145752573","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
SAMHD1 is a human deoxyribonucleoside triphosphatase (dNTPase) known for its role as a restriction factor that targets a wide spectrum of viruses, its involvement in autoimmune disease Aicardi-Goutières syndrome (AGS), and its participation in innate immune regulation. The role of SAMHD1 in cancer, particularly its contribution to drug resistance, has gained increasing attention in recent years. One significant scientific challenge is how to inhibit SAMHD1 function in tumor cells while preserving its function in normal primary cells. Herein, we identified that increased SAMHD1 expression levels correlate with poor prognosis across multiple cancer types, and that SAMHD1 is upregulated in a variety of tumors. Through proteomic analysis and drug screening, we identified a promising strategy for selectively depleting tumor-associated SAMHD1 while minimizing its impact on SAMHD1 expression in key normal cell types. Our approach effectively enhanced tumor cytotoxicity when combined with multiple chemotherapeutic agents and suppressed tumor growth in vivo. Moreover, selective depletion of tumor-associated SAMHD1 activated innate immune responses, leading to enhanced tumor cell killing by immune cells. Collectively, these findings suggest that targeting tumor-specific SAMHD1 represents a novel and promising therapeutic strategy for cancers characterized by elevated SAMHD1 expression, offering potential for improved treatment outcomes in cancer patients with high SAMHD1 expression.
{"title":"Selective depletion of tumor-associated SAMHD1 enhances chemotherapeutic efficacy and antitumor immune responses.","authors":"Jing Sun,Wenwen Zheng,Zheng-Guo Zhang,Hongkun Zhou,Songdi Wang,Dingbo Huang,Xiao-Yi Hu,Qing-Feng Yu,Zhao-Xing Wu,Yi-Fei Shi,Runxin Ye,Fengyan Xia,Wangwei Li,Shurui Lyu,Yu Huang,Xu-Zhao Zhang,Fei Xu,Ke Zhao,Jie Yang,Juan Du,Jiaming Su,Yajuan Rui,Rongzhen Xu,Wei-Ming Yang,Cang Li,Jia Ling Xu,Ruiyu Zhu,Xiaoguang Wang,Wei Wei,Xiao-Fang Yu","doi":"10.1038/s41392-025-02523-1","DOIUrl":"https://doi.org/10.1038/s41392-025-02523-1","url":null,"abstract":"SAMHD1 is a human deoxyribonucleoside triphosphatase (dNTPase) known for its role as a restriction factor that targets a wide spectrum of viruses, its involvement in autoimmune disease Aicardi-Goutières syndrome (AGS), and its participation in innate immune regulation. The role of SAMHD1 in cancer, particularly its contribution to drug resistance, has gained increasing attention in recent years. One significant scientific challenge is how to inhibit SAMHD1 function in tumor cells while preserving its function in normal primary cells. Herein, we identified that increased SAMHD1 expression levels correlate with poor prognosis across multiple cancer types, and that SAMHD1 is upregulated in a variety of tumors. Through proteomic analysis and drug screening, we identified a promising strategy for selectively depleting tumor-associated SAMHD1 while minimizing its impact on SAMHD1 expression in key normal cell types. Our approach effectively enhanced tumor cytotoxicity when combined with multiple chemotherapeutic agents and suppressed tumor growth in vivo. Moreover, selective depletion of tumor-associated SAMHD1 activated innate immune responses, leading to enhanced tumor cell killing by immune cells. Collectively, these findings suggest that targeting tumor-specific SAMHD1 represents a novel and promising therapeutic strategy for cancers characterized by elevated SAMHD1 expression, offering potential for improved treatment outcomes in cancer patients with high SAMHD1 expression.","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"4 1","pages":"406"},"PeriodicalIF":39.3,"publicationDate":"2025-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145752521","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}