首页 > 最新文献

Drug Resistance Updates最新文献

英文 中文
Loss of cyclin C drives resistance to anti-TIGIT therapy by upregulating CD155-mediated immune evasion 细胞周期蛋白C的缺失通过上调cd155介导的免疫逃避来驱动抗tigit治疗的耐药性
IF 21.7 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-10 DOI: 10.1016/j.drup.2025.101318
Shiyu Mao , Yadong Guo , Chengyuan Dong , Dongdong Wang , Xinbo Wang , Linjun Weng , Yanrong Yang , Yaxu Li , Tingting Niu , Qi Wu , Zening Zheng , Zezhi Shan , Xiao Tan , Yaohui Gao , Jiali Jin , Ping Wang , Xin Ge , Bing Shen , Xudong Yao , Lan Fang

Aims

CD155 is an immune checkpoint protein expressed in tumor cells that interacts with its ligand T cell immunoreceptor with immunoglobulin and ITIM domain (TIGIT) on natural killer (NK) cells and T cells, mediating inhibitory regulation on immune cells. Blockade of the CD155-TIGIT interaction has demonstrated clinical benefits in patients with advanced cancers. The transcriptional and post-translational mechanisms governing CD155 expression remain largely unknown.

Methods

To identify regulators of CD155, we conducted a genome-wide CRISPR-Cas9 screen in cancer cells. Surface CD155 protein levels were analyzed via flow cytometry. The role of candidate regulators was validated through loss- and gain-of-function experiments with flow cytometry, Western blot, quantitative PCR, and chromatin immunoprecipitation (ChIP) assays. Additionally, ubiquitination assay was performed to examine post-translational modifications. Functional studies, including NK and T cell cytotoxicity assays, were conducted to assess the immune modulatory effects of CD155 regulation. Clinical relevance was evaluated by analyzing Cyclin C (CCNC) and CD155 expression in datasets of cancer patients who underwent immune checkpoint blockade therapy.

Results

The CRISPR-Cas9 screen identified CCNC as a transcriptional suppressor of CD155. CCNC knockout led to increased surface CD155 expression in cancer cell lines. Mechanistically, CCNC inhibited CD155 transcription by suppressing the activity of the transcription factor FOSL2. Furthermore, CCNC was found to be ubiquitinated and degraded by the E3 ubiquitin ligase FBXO11, suggesting a post-translational regulatory mechanism. Functionally, loss of CCNC promoted CD155 upregulation, thereby enhancing tumor immune evasion from NK and T cell-mediated responses. Clinically, CCNC expression was negatively correlated with CD155 levels in cancer patients, particularly those receiving immune checkpoint blockade therapy.

Conclusion

This study identifies a previously unrecognized master regulator CCNC that functions as a suppressor of CD155-mediated cancer immune evasion. The findings of this study suggest that tumors with low CCNC expression may be resistant to monotherapy and highlight a combination immunotherapy (TIGIT/PD-1 co-blockade) as a promising anti-cancer therapeutic strategy to overcome immune evasion in CCNC-deficient tumors.
AimsCD155是一种在肿瘤细胞中表达的免疫检查点蛋白,它与自然杀伤细胞(NK)和T细胞上的配体T细胞免疫受体与免疫球蛋白和ITIM结构域(TIGIT)相互作用,介导对免疫细胞的抑制调节。阻断CD155-TIGIT相互作用已证明对晚期癌症患者有临床益处。调控CD155表达的转录和翻译后机制在很大程度上仍然未知。方法为了鉴定CD155的调控因子,我们在癌细胞中进行了全基因组CRISPR-Cas9筛选。流式细胞术分析表面CD155蛋白水平。候选调节因子的作用通过流式细胞术、Western blot、定量PCR和染色质免疫沉淀(ChIP)测定的功能缺失和功能获得实验得到验证。此外,通过泛素化实验检测翻译后修饰。功能研究,包括NK和T细胞毒性试验,被用于评估CD155调控的免疫调节作用。通过分析接受免疫检查点阻断治疗的癌症患者数据集中的细胞周期蛋白C (CCNC)和CD155表达来评估临床相关性。结果CRISPR-Cas9筛选鉴定出CCNC是CD155的转录抑制因子。敲除CCNC导致癌细胞株表面CD155表达增加。在机制上,CCNC通过抑制转录因子FOSL2的活性来抑制CD155的转录。此外,发现CCNC被E3泛素连接酶FBXO11泛素化和降解,提示翻译后调控机制。在功能上,CCNC的缺失促进了CD155的上调,从而增强了NK和T细胞介导的肿瘤免疫逃避反应。在临床上,癌症患者,特别是接受免疫检查点阻断治疗的患者,CCNC表达与CD155水平呈负相关。本研究确定了一个以前未被识别的主调控因子CCNC,其功能是cd155介导的癌症免疫逃避的抑制因子。本研究结果表明,低CCNC表达的肿瘤可能对单一治疗有耐药性,并强调联合免疫治疗(TIGIT/PD-1共阻断)是一种有希望的抗癌治疗策略,可以克服CCNC缺陷肿瘤的免疫逃避。
{"title":"Loss of cyclin C drives resistance to anti-TIGIT therapy by upregulating CD155-mediated immune evasion","authors":"Shiyu Mao ,&nbsp;Yadong Guo ,&nbsp;Chengyuan Dong ,&nbsp;Dongdong Wang ,&nbsp;Xinbo Wang ,&nbsp;Linjun Weng ,&nbsp;Yanrong Yang ,&nbsp;Yaxu Li ,&nbsp;Tingting Niu ,&nbsp;Qi Wu ,&nbsp;Zening Zheng ,&nbsp;Zezhi Shan ,&nbsp;Xiao Tan ,&nbsp;Yaohui Gao ,&nbsp;Jiali Jin ,&nbsp;Ping Wang ,&nbsp;Xin Ge ,&nbsp;Bing Shen ,&nbsp;Xudong Yao ,&nbsp;Lan Fang","doi":"10.1016/j.drup.2025.101318","DOIUrl":"10.1016/j.drup.2025.101318","url":null,"abstract":"<div><h3>Aims</h3><div>CD155 is an immune checkpoint protein expressed in tumor cells that interacts with its ligand T cell immunoreceptor with immunoglobulin and ITIM domain (TIGIT) on natural killer (NK) cells and T cells, mediating inhibitory regulation on immune cells. Blockade of the CD155-TIGIT interaction has demonstrated clinical benefits in patients with advanced cancers. The transcriptional and post-translational mechanisms governing CD155 expression remain largely unknown.</div></div><div><h3>Methods</h3><div>To identify regulators of CD155, we conducted a genome-wide CRISPR-Cas9 screen in cancer cells. Surface CD155 protein levels were analyzed via flow cytometry. The role of candidate regulators was validated through loss- and gain-of-function experiments with flow cytometry, Western blot, quantitative PCR, and chromatin immunoprecipitation (ChIP) assays. Additionally, ubiquitination assay was performed to examine post-translational modifications. Functional studies, including NK and T cell cytotoxicity assays, were conducted to assess the immune modulatory effects of CD155 regulation. Clinical relevance was evaluated by analyzing Cyclin C (CCNC) and CD155 expression in datasets of cancer patients who underwent immune checkpoint blockade therapy.</div></div><div><h3>Results</h3><div>The CRISPR-Cas9 screen identified CCNC as a transcriptional suppressor of CD155. <em>CCNC</em> knockout led to increased surface CD155 expression in cancer cell lines. Mechanistically, CCNC inhibited CD155 transcription by suppressing the activity of the transcription factor FOSL2. Furthermore, CCNC was found to be ubiquitinated and degraded by the E3 ubiquitin ligase FBXO11, suggesting a post-translational regulatory mechanism. Functionally, loss of CCNC promoted CD155 upregulation, thereby enhancing tumor immune evasion from NK and T cell-mediated responses. Clinically, CCNC expression was negatively correlated with CD155 levels in cancer patients, particularly those receiving immune checkpoint blockade therapy.</div></div><div><h3>Conclusion</h3><div>This study identifies a previously unrecognized master regulator CCNC that functions as a suppressor of CD155-mediated cancer immune evasion. The findings of this study suggest that tumors with low CCNC expression may be resistant to monotherapy and highlight a combination immunotherapy (TIGIT/PD-1 co-blockade) as a promising anti-cancer therapeutic strategy to overcome immune evasion in CCNC-deficient tumors.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"84 ","pages":"Article 101318"},"PeriodicalIF":21.7,"publicationDate":"2025-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145485479","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}
引用次数: 0
Y10 phosphorylation of LDHA promotes the release of extracellular vesicle-derived circSEPT9 to enhance the chemoresistance of triple negative breast cancer cells via modulation of miR-515–5p/KIAA1429 axis Y10磷酸化LDHA促进细胞外囊泡衍生的circSEPT9的释放,通过调节miR-515-5p/KIAA1429轴增强三阴性乳腺癌细胞的化疗耐药
IF 21.7 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-07 DOI: 10.1016/j.drup.2025.101324
Yueping Wang , Ziyun Zhang , Juan Gu , Ming Zhou , Jiankang Huang , Daoping Zhou , Xuedong Wang

Objective

Extracellular vesicle (EV)-derived RNAs play crucial roles in cancer biology and therapeutic resistance. This study investigated how EV-derived circSEPT9 mediates doxorubicin (DOX) resistance in triple negative breast cancer (TNBC).

Methods

TNBC tissues and samples were analyzed alongside MDA-MB-231 and MDA-MB-231/ADR cells. The molecular interplay among circSEPT9, KIAA1429, and miR-515–5p was explored to elucidate the regulatory axis underlying drug resistance. EVs were purified to assess the clinical diagnostic potential of EV-circSEPT9. Functional assays employing lactate dehydrogenase A (LDHA) knockdown and rescue with Flag-tagged human LDHA WT or Y10F mutant constructs were conducted to examine the significance of LDHA Y10 phosphorylation in EV-circSEPT9 release and DOX resistance.

Results

circSEPT9 silencing impaired proliferation, invasion, and colony formation of DOX-resistant TNBC cells while increasing their sensitivity to DOX through suppression of KIAA1429-mediated m6A modification. EVs from resistant cells transferred circSEPT9 to sensitive cells, thereby enhancing their drug resistance and tumor growth capacity. LDHA Y10 phosphorylation was found to be indispensable for EV-circSEPT9 secretion and the subsequent intercellular transfer of DOX resistance.

Conclusion

Phosphorylated LDHA (Y10) promotes EV-circSEPT9 secretion, elevating intracellular circSEPT9 levels in recipient TNBC cells. By functioning as a competing endogenous RNA (ceRNA) that sponges miR-515–5p, circSEPT9 upregulates KIAA1429, augments m6A methylation, and drives the development of chemoresistance.
目的细胞外囊泡(EV)来源的rna在肿瘤生物学和治疗耐药中发挥重要作用。本研究探讨ev衍生的circSEPT9如何介导三阴性乳腺癌(TNBC)的阿霉素(DOX)耐药。方法用MDA-MB-231和MDA-MB-231/ADR细胞对stnbc组织和样本进行分析。我们探索了circSEPT9、KIAA1429和miR-515-5p之间的分子相互作用,以阐明耐药背后的调控轴。对ev进行纯化以评估EV-circSEPT9的临床诊断潜力。采用乳酸脱氢酶A (LDHA)敲除和用flag标记的人LDHA WT或Y10F突变体构建挽救进行功能分析,以检验LDHA Y10磷酸化在EV-circSEPT9释放和DOX抗性中的意义。结果scircsept9通过抑制kiaa1429介导的m6A修饰,抑制了DOX耐药TNBC细胞的增殖、侵袭和集落形成,同时增加了它们对DOX的敏感性。来自耐药细胞的ev将circSEPT9转移到敏感细胞,从而增强其耐药性和肿瘤生长能力。发现LDHA Y10磷酸化对于EV-circSEPT9的分泌和随后的DOX抗性的细胞间转移是必不可少的。结论磷酸化的LDHA (Y10)促进EV-circSEPT9分泌,升高受体TNBC细胞内circSEPT9水平。circSEPT9作为一种竞争的内源性RNA (ceRNA),可以吸收miR-515-5p,从而上调KIAA1429,增强m6A甲基化,并推动化学耐药的发展。
{"title":"Y10 phosphorylation of LDHA promotes the release of extracellular vesicle-derived circSEPT9 to enhance the chemoresistance of triple negative breast cancer cells via modulation of miR-515–5p/KIAA1429 axis","authors":"Yueping Wang ,&nbsp;Ziyun Zhang ,&nbsp;Juan Gu ,&nbsp;Ming Zhou ,&nbsp;Jiankang Huang ,&nbsp;Daoping Zhou ,&nbsp;Xuedong Wang","doi":"10.1016/j.drup.2025.101324","DOIUrl":"10.1016/j.drup.2025.101324","url":null,"abstract":"<div><h3>Objective</h3><div>Extracellular vesicle (EV)-derived RNAs play crucial roles in cancer biology and therapeutic resistance. This study investigated how EV-derived circSEPT9 mediates doxorubicin (DOX) resistance in triple negative breast cancer (TNBC).</div></div><div><h3>Methods</h3><div>TNBC tissues and samples were analyzed alongside MDA-MB-231 and MDA-MB-231/ADR cells. The molecular interplay among circSEPT9, KIAA1429, and miR-515–5p was explored to elucidate the regulatory axis underlying drug resistance. EVs were purified to assess the clinical diagnostic potential of EV-circSEPT9. Functional assays employing lactate dehydrogenase A (LDHA) knockdown and rescue with Flag-tagged human LDHA WT or Y10F mutant constructs were conducted to examine the significance of LDHA Y10 phosphorylation in EV-circSEPT9 release and DOX resistance.</div></div><div><h3>Results</h3><div>circSEPT9 silencing impaired proliferation, invasion, and colony formation of DOX-resistant TNBC cells while increasing their sensitivity to DOX through suppression of KIAA1429-mediated m6A modification. EVs from resistant cells transferred circSEPT9 to sensitive cells, thereby enhancing their drug resistance and tumor growth capacity. LDHA Y10 phosphorylation was found to be indispensable for EV-circSEPT9 secretion and the subsequent intercellular transfer of DOX resistance.</div></div><div><h3>Conclusion</h3><div>Phosphorylated LDHA (Y10) promotes EV-circSEPT9 secretion, elevating intracellular circSEPT9 levels in recipient TNBC cells. By functioning as a competing endogenous RNA (ceRNA) that sponges miR-515–5p, circSEPT9 upregulates KIAA1429, augments m6A methylation, and drives the development of chemoresistance.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"84 ","pages":"Article 101324"},"PeriodicalIF":21.7,"publicationDate":"2025-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145462288","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}
引用次数: 0
Metabolite-driven reprogramming of bacterial persisters: Mechanisms and therapeutic opportunities for overcoming antibiotic tolerance 代谢物驱动的细菌持久性重编程:克服抗生素耐受性的机制和治疗机会
IF 21.7 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-05 DOI: 10.1016/j.drup.2025.101322
Yixiao Song , Zifan Ye , Yipeng Wang
Bacterial persisters constitute a heterogeneous subpopulation of dormant or slow-growing cells capable of surviving harsh environmental conditions, including antibiotic exposure. These cells are strongly associated with the failure of clinical antibiotic therapies and the recurrence of chronic bacterial infections. A comprehensive understanding of the physiological features of bacterial persisters is therefore critical for the development of targeted interventions aimed at overcoming the limitations of conventional antibiotic treatments. In this review, we summarize current models of bacterial persister formation, with particular emphasis on the pivotal role of reduced metabolic activity in mediating antibiotic tolerance. We further elucidate the mechanisms through which exogenous metabolites—such as sugars, lipids, and nucleic acid derivatives—induce metabolic reprogramming, thereby reversing multidrug resistance in bacterial persisters. Furthermore, we summarized the major barriers limiting the clinical translation of metabolite-assisted “wake-and-kill” strategies and outlined future prospects for their application. In conclusion, restoring bacterial metabolic activity through targeted metabolite interventions represents a promising avenue to overcome antibiotic tolerance, paving the way for next-generation therapeutic strategies against persistent infections.
细菌持久性构成了一种异质性的休眠或生长缓慢的细胞亚群,能够在恶劣的环境条件下生存,包括抗生素暴露。这些细胞与临床抗生素治疗的失败和慢性细菌感染的复发密切相关。因此,全面了解细菌持续存在的生理特征对于开发旨在克服传统抗生素治疗局限性的针对性干预措施至关重要。在这篇综述中,我们总结了目前细菌持久性形成的模型,特别强调了代谢活性降低在介导抗生素耐受性中的关键作用。我们进一步阐明了外源性代谢物(如糖、脂质和核酸衍生物)诱导代谢重编程的机制,从而逆转了细菌持久性的多药耐药。此外,我们总结了限制代谢物辅助“唤醒和杀死”策略临床转化的主要障碍,并概述了其应用的未来前景。总之,通过有针对性的代谢物干预来恢复细菌的代谢活性是克服抗生素耐受性的一个有希望的途径,为针对持续感染的下一代治疗策略铺平了道路。
{"title":"Metabolite-driven reprogramming of bacterial persisters: Mechanisms and therapeutic opportunities for overcoming antibiotic tolerance","authors":"Yixiao Song ,&nbsp;Zifan Ye ,&nbsp;Yipeng Wang","doi":"10.1016/j.drup.2025.101322","DOIUrl":"10.1016/j.drup.2025.101322","url":null,"abstract":"<div><div>Bacterial persisters constitute a heterogeneous subpopulation of dormant or slow-growing cells capable of surviving harsh environmental conditions, including antibiotic exposure. These cells are strongly associated with the failure of clinical antibiotic therapies and the recurrence of chronic bacterial infections. A comprehensive understanding of the physiological features of bacterial persisters is therefore critical for the development of targeted interventions aimed at overcoming the limitations of conventional antibiotic treatments. In this review, we summarize current models of bacterial persister formation, with particular emphasis on the pivotal role of reduced metabolic activity in mediating antibiotic tolerance. We further elucidate the mechanisms through which exogenous metabolites—such as sugars, lipids, and nucleic acid derivatives—induce metabolic reprogramming, thereby reversing multidrug resistance in bacterial persisters. Furthermore, we summarized the major barriers limiting the clinical translation of metabolite-assisted “wake-and-kill” strategies and outlined future prospects for their application. In conclusion, restoring bacterial metabolic activity through targeted metabolite interventions represents a promising avenue to overcome antibiotic tolerance, paving the way for next-generation therapeutic strategies against persistent infections.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"84 ","pages":"Article 101322"},"PeriodicalIF":21.7,"publicationDate":"2025-11-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145441918","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}
引用次数: 0
Nanoparticles-mediated mitochondrial relocation of lipid-lowering drugs shape energy metabolism to conquer acquired immune resistance 纳米颗粒介导的降脂药物线粒体重新定位塑造能量代谢以克服获得性免疫抵抗
IF 21.7 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-04 DOI: 10.1016/j.drup.2025.101323
Cheng Li , Wei Xiong , Jiahao Liu , Ke Li , Haoxiang Wang , Zhengxiang Wang , Feiyu Liu , Jianliang Shen , Zaigang Zhou , Shenpeng Ying , Long Wang
CD276, is a fatal recently discovered immune checkpoint proteins of B7 family. Due to the not clearly uncovered signal pathways that involved in the expression of CD276 in tumors, few strategies were discovered to regulate CD276. Here, we newly discovered that abnormal tumor mitochondrial activation played a vital important role in raising CD276 expression through targeting AMPK/mTOR signal pathway. Then, it was also revealed that clinical usable lipid-lowering drugs with mitochondria oxidative phosphorylation (OXPHOS) and glycolysis inhibiting capacity, like fenofibric acid (FFA), exhibited desired programmed death ligand-1 (PD-L1) and CD276 co-suppression capacity. To better deliver FFA to tumor mitochondria, IR-FFA was synthesized by linking the mitochondria-targeting heptamethylene cyanine IR-68 with FFA, followed by self-assembly with albumin (Alb) to create IR-FFA@Alb nanoparticles. By doing so, the dosage needed for IR-FFA@Alb to depress CD276 and PD-L1 expression was 100 times lower than free FFA. Then, IR-FFA@Alb monotherapy effectively inhibited tumor growth both in vitro and in vivo. Moreover, the combination therapy of IR-FFA@Alb nanoparticles and radiotherapy (RT) effectively avoid the frequently occurred immune tolerance phenomenon of RT by co-depression CD276 and PD-L1. These results altogether showed the possibility of using lipid-lowering drugs as multi-functional immune checkpoint inhibitors to sensitize tumor therapy.
CD276是最近发现的一种致命的B7家族免疫检查点蛋白。由于肿瘤中参与CD276表达的信号通路尚未被明确发现,因此很少发现调节CD276的策略。本研究中新发现,肿瘤线粒体异常激活通过靶向AMPK/mTOR信号通路,在上调CD276表达中起着至关重要的作用。然后,还揭示了临床可用的具有线粒体氧化磷酸化(OXPHOS)和糖酵解抑制能力的降脂药物,如非诺纤维酸(FFA),表现出所需的程序性死亡配体-1 (PD-L1)和CD276共抑制能力。为了更好地将FFA传递到肿瘤线粒体,将靶向线粒体的七亚甲基菁氨酸IR-68与FFA连接,然后与白蛋白(Alb)自组装,合成IR-FFA纳米颗粒IR-FFA@Alb。通过这样做,IR-FFA@Alb抑制CD276和PD-L1表达所需的剂量比游离FFA低100倍。然后,IR-FFA@Alb单药治疗有效地抑制了肿瘤的体外和体内生长。此外,IR-FFA@Alb纳米颗粒与放疗(RT)联合治疗通过共同抑制CD276和PD-L1,有效避免了RT中经常发生的免疫耐受现象。这些结果共同表明,使用降脂药物作为多功能免疫检查点抑制剂来增敏肿瘤治疗的可能性。
{"title":"Nanoparticles-mediated mitochondrial relocation of lipid-lowering drugs shape energy metabolism to conquer acquired immune resistance","authors":"Cheng Li ,&nbsp;Wei Xiong ,&nbsp;Jiahao Liu ,&nbsp;Ke Li ,&nbsp;Haoxiang Wang ,&nbsp;Zhengxiang Wang ,&nbsp;Feiyu Liu ,&nbsp;Jianliang Shen ,&nbsp;Zaigang Zhou ,&nbsp;Shenpeng Ying ,&nbsp;Long Wang","doi":"10.1016/j.drup.2025.101323","DOIUrl":"10.1016/j.drup.2025.101323","url":null,"abstract":"<div><div>CD276, is a fatal recently discovered immune checkpoint proteins of B7 family. Due to the not clearly uncovered signal pathways that involved in the expression of CD276 in tumors, few strategies were discovered to regulate CD276. Here, we newly discovered that abnormal tumor mitochondrial activation played a vital important role in raising CD276 expression through targeting AMPK/mTOR signal pathway. Then, it was also revealed that clinical usable lipid-lowering drugs with mitochondria oxidative phosphorylation (OXPHOS) and glycolysis inhibiting capacity, like fenofibric acid (FFA), exhibited desired programmed death ligand-1 (PD-L1) and CD276 co-suppression capacity. To better deliver FFA to tumor mitochondria, IR-FFA was synthesized by linking the mitochondria-targeting heptamethylene cyanine IR-68 with FFA, followed by self-assembly with albumin (Alb) to create IR-FFA@Alb nanoparticles. By doing so, the dosage needed for IR-FFA@Alb to depress CD276 and PD-L1 expression was 100 times lower than free FFA. Then, IR-FFA@Alb monotherapy effectively inhibited tumor growth both <em>in vitro</em> and <em>in vivo</em>. Moreover, the combination therapy of IR-FFA@Alb nanoparticles and radiotherapy (RT) effectively avoid the frequently occurred immune tolerance phenomenon of RT by co-depression CD276 and PD-L1. These results altogether showed the possibility of using lipid-lowering drugs as multi-functional immune checkpoint inhibitors to sensitize tumor therapy.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"84 ","pages":"Article 101323"},"PeriodicalIF":21.7,"publicationDate":"2025-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145441447","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}
引用次数: 0
Disruption of NANOG-driven epithelial-mesenchymal transition (EMT) and self-renewal restores drug sensitivity in colorectal cancer 破坏nanog驱动的上皮-间质转化(EMT)和自我更新可恢复结直肠癌的药物敏感性
IF 21.7 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-04 DOI: 10.1016/j.drup.2025.101321
Kiarash Saleki , Sameerah Shaheen , Miao Xue , Amirreza Mazloomi , Sepideh Youssefi , Hossein Kashfi , Mehreen Ahmed , Roya Babaei-Jadidi , Bradley Spencer-Dene , Dominique Bonnet , Chris Denning , Abdolrahman S. Nateri

Aims

To investigate the regulatory role of NANOG in genes associated with stemness, symmetric division, and therapeutic resistance in colorectal cancer stem-like cells (CRC-SCs), with a focus on ERK/GSK-3β/β-catenin signalling and epithelial-mesenchymal transition (EMT), in order to evaluate the translational potential of targeting NANOG-associated signalling pathways.

Methods

Stemness, signalling activity, and cell division modes were analysed using 3D colonospheres enriched for CRC-SCs. Drug responses to the MEK inhibitor U0126 and the GSK-3β inhibitor TDZD-8 were assessed in CRC patient-derived organoids (PDOs), alongside molecular assays, immunohistochemistry with H-score quantification in xenograft models, and molecular dynamics simulations.

Results

NANOG overexpression enhanced the expression of stemness-associated genes, promoted symmetric cell division, and activated ERK/GSK-3β signalling, contributing to increased sphere formation. Inhibition of MEK and GSK-3β reduced EMT, cell proliferation, and symmetric division in CRC-SCs. NANOG-mediated dysregulation of ERK/GSK-3β altered β-catenin signalling and disrupted E-cadherin-dependent cell-cell adhesion. Molecular simulations and drug assays demonstrated that TDZD-8 and U0126 interfere with NANOG-DNA binding and β-catenin/E-cadherin interactions.

Conclusions

NANOG drives CRC-SC maintenance via ERK/GSK-3β/β-catenin signalling and EMT modulation. This study offers significant insights into the translational impact of targeting NANOG and its downstream pathways with small-molecule inhibitors U0126 and TDZD-8 and presents a promising strategy to reduce CRC-SCs stemness, functionality, and tumourigenicity.
目的研究NANOG在结直肠癌干细胞样细胞(CRC-SCs)干性、对称分裂和治疗耐药相关基因中的调控作用,重点研究ERK/GSK-3β/β-catenin信号传导和上皮-间质转化(EMT),以评估靶向NANOG相关信号通路的翻译潜力。方法利用富含CRC-SCs的3D结肠球分析细胞的系统性、信号活性和细胞分裂模式。在结直肠癌患者衍生类器官(PDOs)中评估MEK抑制剂U0126和GSK-3β抑制剂TDZD-8的药物反应,同时进行分子分析、异种移植模型中h评分定量的免疫组织化学和分子动力学模拟。结果snanog过表达可增强干细胞相关基因的表达,促进细胞对称分裂,激活ERK/GSK-3β信号通路,促进球的形成。抑制MEK和GSK-3β可降低CRC-SCs的EMT、细胞增殖和对称分裂。nanog介导的ERK/GSK-3β的失调改变了β-catenin信号传导并破坏了e -cadherin依赖性细胞-细胞粘附。分子模拟和药物实验表明,TDZD-8和U0126干扰NANOG-DNA结合和β-catenin/E-cadherin相互作用。结论snanog通过ERK/GSK-3β/β-catenin信号传导和EMT调控驱动CRC-SC维持。该研究为小分子抑制剂U0126和TDZD-8靶向NANOG及其下游通路的翻译影响提供了重要见解,并提出了降低CRC-SCs的干细胞性、功能和致瘤性的有希望的策略。
{"title":"Disruption of NANOG-driven epithelial-mesenchymal transition (EMT) and self-renewal restores drug sensitivity in colorectal cancer","authors":"Kiarash Saleki ,&nbsp;Sameerah Shaheen ,&nbsp;Miao Xue ,&nbsp;Amirreza Mazloomi ,&nbsp;Sepideh Youssefi ,&nbsp;Hossein Kashfi ,&nbsp;Mehreen Ahmed ,&nbsp;Roya Babaei-Jadidi ,&nbsp;Bradley Spencer-Dene ,&nbsp;Dominique Bonnet ,&nbsp;Chris Denning ,&nbsp;Abdolrahman S. Nateri","doi":"10.1016/j.drup.2025.101321","DOIUrl":"10.1016/j.drup.2025.101321","url":null,"abstract":"<div><h3>Aims</h3><div>To investigate the regulatory role of NANOG in genes associated with stemness, symmetric division, and therapeutic resistance in colorectal cancer stem-like cells (CRC-SCs), with a focus on ERK/GSK-3β/β-catenin signalling and epithelial-mesenchymal transition (EMT), in order to evaluate the translational potential of targeting NANOG-associated signalling pathways.</div></div><div><h3>Methods</h3><div>Stemness, signalling activity, and cell division modes were analysed using 3D colonospheres enriched for CRC-SCs. Drug responses to the MEK inhibitor U0126 and the GSK-3β inhibitor TDZD-8 were assessed in CRC patient-derived organoids (PDOs), alongside molecular assays, immunohistochemistry with H-score quantification in xenograft models, and molecular dynamics simulations.</div></div><div><h3>Results</h3><div>NANOG overexpression enhanced the expression of stemness-associated genes, promoted symmetric cell division, and activated ERK/GSK-3β signalling, contributing to increased sphere formation. Inhibition of MEK and GSK-3β reduced EMT, cell proliferation, and symmetric division in CRC-SCs. NANOG-mediated dysregulation of ERK/GSK-3β altered β-catenin signalling and disrupted E-cadherin-dependent cell-cell adhesion. Molecular simulations and drug assays demonstrated that TDZD-8 and U0126 interfere with NANOG-DNA binding and β-catenin/E-cadherin interactions.</div></div><div><h3>Conclusions</h3><div>NANOG drives CRC-SC maintenance via ERK/GSK-3β/β-catenin signalling and EMT modulation. This study offers significant insights into the translational impact of targeting NANOG and its downstream pathways with small-molecule inhibitors U0126 and TDZD-8 and presents a promising strategy to reduce CRC-SCs stemness, functionality, and tumourigenicity.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"84 ","pages":"Article 101321"},"PeriodicalIF":21.7,"publicationDate":"2025-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145441940","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}
引用次数: 0
The role of synthetic lethality in overcoming cancer therapy resistance: Emerging paradigm and recent advances. 合成致死率在克服癌症治疗耐药中的作用:新兴范式和最新进展。
IF 21.7 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-01 Epub Date: 2025-08-07 DOI: 10.1016/j.drup.2025.101290
Qingyi Xiong, Jinmei Jin, Jiayi Lin, Bohan Zhang, Yixin Jiang, Zhe Sun, Lijun Zhang, Ye Wu, Guozhi Zhao, Jiang-Jiang Qin, Xin Luan

Cancer therapy resistance remains a major barrier to successful treatment, often leading to reduced clinical efficacy or cancer relapse. Synthetic lethality (SL) has emerged as a promising strategy to exploit genetic vulnerabilities in cancer cells, allowing for more selective and less toxic therapies. By leveraging the genetic or non-genetic adaptations that cancer cells develop under therapeutic pressure, SL-based therapies provide a more precise and less toxic treatment approach. Additionally, SL-driven drug combinations not only delay development of drug resistance but also enhance therapeutic efficacy, representing a transformative shift in cancer management. A comprehensive understanding of SL mechanisms in the context of drug resistance is essential for advancing effective treatment strategies. This review highlights recent advances in SL research, emphasizing the gene screening techniques in overcoming cancer therapy resistance.

癌症治疗耐药性仍然是成功治疗的主要障碍,往往导致临床疗效降低或癌症复发。合成致死性(SL)已成为利用癌细胞遗传脆弱性的一种很有前途的策略,允许更有选择性和更少毒性的治疗。通过利用癌细胞在治疗压力下产生的遗传或非遗传适应性,基于sl的治疗提供了一种更精确、毒性更小的治疗方法。此外,sl驱动的药物组合不仅延缓了耐药性的发展,而且提高了治疗效果,代表了癌症管理的革命性转变。在耐药背景下,全面了解SL机制对于推进有效的治疗策略至关重要。本文综述了SL研究的最新进展,重点介绍了基因筛选技术在克服肿瘤治疗耐药中的应用。
{"title":"The role of synthetic lethality in overcoming cancer therapy resistance: Emerging paradigm and recent advances.","authors":"Qingyi Xiong, Jinmei Jin, Jiayi Lin, Bohan Zhang, Yixin Jiang, Zhe Sun, Lijun Zhang, Ye Wu, Guozhi Zhao, Jiang-Jiang Qin, Xin Luan","doi":"10.1016/j.drup.2025.101290","DOIUrl":"10.1016/j.drup.2025.101290","url":null,"abstract":"<p><p>Cancer therapy resistance remains a major barrier to successful treatment, often leading to reduced clinical efficacy or cancer relapse. Synthetic lethality (SL) has emerged as a promising strategy to exploit genetic vulnerabilities in cancer cells, allowing for more selective and less toxic therapies. By leveraging the genetic or non-genetic adaptations that cancer cells develop under therapeutic pressure, SL-based therapies provide a more precise and less toxic treatment approach. Additionally, SL-driven drug combinations not only delay development of drug resistance but also enhance therapeutic efficacy, representing a transformative shift in cancer management. A comprehensive understanding of SL mechanisms in the context of drug resistance is essential for advancing effective treatment strategies. This review highlights recent advances in SL research, emphasizing the gene screening techniques in overcoming cancer therapy resistance.</p>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"83 ","pages":"101290"},"PeriodicalIF":21.7,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144812629","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}
引用次数: 0
Peptide nanonet trapping suppresses bacterial motility and delays antibiotic resistance emergence 肽纳米捕获抑制细菌运动和延迟抗生素耐药性的出现
IF 21.7 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-10-29 DOI: 10.1016/j.drup.2025.101320
Jian Xu , Nhan Dai Thien Tram , Peiyan Yu, Dhanya Mahalakshmi Murali, Wei Meng Chen, Samantha Jinglin Yang , Pui Lai Rachel Ee

Aims

In the presence of antibiotics, motile bacteria can navigate chemical gradients for adaptation and survival. Antimicrobial peptides (AMPs) have been widely explored as adjuvant to improve the activity potency of antibiotics, but mainly through the disruption of bacterial membranes. In this work, we investigated the impact of nanonet trapping using fibrillating peptides, a mechanistically unique sub-group of AMPs, on motility recovery of bacteria and their capacity to develop antibiotic resistance.

Methods

The ability of fibrillating AMPs to potentiate activity and delay resistance of antibiotics from diverse classes was evaluated against clinical isolates of Gram-negative pathogens. Using soft agar assay and live-tracking microscopy, shifts in the motility of the bacteria population subjected to different treatments were evaluated. To further elucidate the mechanism of action, the expression of major flagella-encoding genes was quantified and hypomotile bacteria strains were studied.

Results

At sub-inhibitory concentrations, fibrillating peptides not only displayed synergistic interactions, but also significantly delayed the emergence of resistance to antibiotics such as rifampicin for at least 18 days. The peptide-antibiotic synergy profiles were lost after prolonged treatment with antibiotic monotherapy but preserved when co-administered with fibrillating peptides throughout the serial passage. The nanonet-forming peptides were shown to serve as a motility filter where the bacteria population gradually shifted towards homogeneous hypomotility associated with inferior survivability.

Conclusions

This work showcases the potential of AMPs as low-concentration adjuvants for extending the clinical lifespan of current antibiotics and highlights bacterial motility as an underexplored target for antibiotic development.
目的在抗生素的存在下,活动细菌可以通过化学梯度来适应和生存。抗菌肽(Antimicrobial peptides, AMPs)作为提高抗生素活性效力的佐剂已被广泛研究,但主要是通过破坏细菌膜。在这项工作中,我们研究了利用纤颤肽(amp的一个独特的亚群)捕获纳米网络对细菌运动恢复及其产生抗生素耐药性的能力的影响。方法以临床分离的革兰氏阴性病原菌为对照,评价纤颤amp增强不同种类抗生素的活性和延迟耐药性的能力。使用软琼脂实验和实时跟踪显微镜,在不同的处理下,细菌群体的运动能力的变化进行了评估。为了进一步阐明其作用机制,我们对主要鞭毛编码基因的表达进行了量化,并对低动菌菌株进行了研究。结果在亚抑制浓度下,纤颤肽不仅表现出协同作用,而且显著延缓了利福平等抗生素耐药的出现至少18天。在长时间的抗生素单药治疗后,肽-抗生素协同作用谱丢失,但在整个序列通道中与纤颤肽共同施用时保留。纳米形成肽被证明可以作为一种运动过滤器,在那里细菌群体逐渐转向均匀的低运动,与低生存能力相关。结论:本研究显示了抗菌肽作为低浓度佐剂的潜力,可以延长当前抗生素的临床使用寿命,并强调了细菌运动是抗生素开发中一个未被充分开发的靶点。
{"title":"Peptide nanonet trapping suppresses bacterial motility and delays antibiotic resistance emergence","authors":"Jian Xu ,&nbsp;Nhan Dai Thien Tram ,&nbsp;Peiyan Yu,&nbsp;Dhanya Mahalakshmi Murali,&nbsp;Wei Meng Chen,&nbsp;Samantha Jinglin Yang ,&nbsp;Pui Lai Rachel Ee","doi":"10.1016/j.drup.2025.101320","DOIUrl":"10.1016/j.drup.2025.101320","url":null,"abstract":"<div><h3>Aims</h3><div>In the presence of antibiotics, motile bacteria can navigate chemical gradients for adaptation and survival. Antimicrobial peptides (AMPs) have been widely explored as adjuvant to improve the activity potency of antibiotics, but mainly through the disruption of bacterial membranes. In this work, we investigated the impact of nanonet trapping using fibrillating peptides, a mechanistically unique sub-group of AMPs, on motility recovery of bacteria and their capacity to develop antibiotic resistance.</div></div><div><h3>Methods</h3><div>The ability of fibrillating AMPs to potentiate activity and delay resistance of antibiotics from diverse classes was evaluated against clinical isolates of Gram-negative pathogens. Using soft agar assay and live-tracking microscopy, shifts in the motility of the bacteria population subjected to different treatments were evaluated. To further elucidate the mechanism of action, the expression of major flagella-encoding genes was quantified and hypomotile bacteria strains were studied.</div></div><div><h3>Results</h3><div>At sub-inhibitory concentrations, fibrillating peptides not only displayed synergistic interactions, but also significantly delayed the emergence of resistance to antibiotics such as rifampicin for at least 18 days. The peptide-antibiotic synergy profiles were lost after prolonged treatment with antibiotic monotherapy but preserved when co-administered with fibrillating peptides throughout the serial passage. The nanonet-forming peptides were shown to serve as a motility filter where the bacteria population gradually shifted towards homogeneous hypomotility associated with inferior survivability.</div></div><div><h3>Conclusions</h3><div>This work showcases the potential of AMPs as low-concentration adjuvants for extending the clinical lifespan of current antibiotics and highlights bacterial motility as an underexplored target for antibiotic development.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"84 ","pages":"Article 101320"},"PeriodicalIF":21.7,"publicationDate":"2025-10-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145382791","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}
引用次数: 0
ZBP1 antagonizes MRE11-mediated DNA end resection and confers synthetic lethality to PARP inhibition in ovarian cancer ZBP1拮抗mre11介导的DNA末端切除,并赋予PARP抑制在卵巢癌中的合成致死性
IF 21.7 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-10-28 DOI: 10.1016/j.drup.2025.101319
Shen-nan Shi , Qiuyang Xu , Zhiqi Liao , Wenjian Gong , Yilin Cui , Jiahao Liu , Xiaofei Jiao , Yijie Wu , Mengshi Luo , Yuewen Zhang , Linghui Wang , Yuanjia Wen , Wen Pan , Xuejiao Zhao , Marilyne Labrie , Zhiyong Ding , Gordon B. Mills , Ding Ma , Guang-Nian Zhao , Qinglei Gao , Yong Fang
ZBP1, a classic pattern recognition receptor (PRR), has been implicated in regulating programmed cell death and the innate immune response. However, the role of ZBP1 in the nucleus remains largely undefined. Here, we found that nuclear ZBP1 localizes to the site of DNA double-stranded breaks (DSBs) following DNA damage and impairs homologous recombination (HR) repair through its interaction with MRE11. ZBP1 interacts with MRE11 through RHIM A and B domains and inhibits the enzymatic activity of MRE11, ultimately leading to the suppression of HR and DNA damage repair (DDR). These processes are initiated via ATM-mediated ZBP1 phosphorylation at S106. Consistent with these findings, in vitro and in vivo models both exhibit increased sensitivity to PARP inhibitor treatment following ZBP1 overexpression. Furthermore, in our neoadjuvant niraparib monotherapy study (NCT05407841) higher ZBP1 expression correlates with better response to PARP inhibition and prolonged PFS in high-grade serous ovarian cancer (HGSOC). This study describes a novel function of ZBP1 for regulating HR, which confers synthetic lethality to PARP inhibition in ovarian cancer. ZBP1 thus serves as a potential therapy target and biomarker of response to PARP inhibitors and potentially other therapeutic agents such as platin analogs that are synthetically lethal with defective HR.
ZBP1是一种典型的模式识别受体(PRR),参与调节程序性细胞死亡和先天免疫反应。然而,ZBP1在细胞核中的作用仍未明确。在这里,我们发现细胞核ZBP1定位于DNA损伤后的DNA双链断裂(DSBs)位点,并通过与MRE11的相互作用损害同源重组(HR)修复。ZBP1通过RHIM A和B结构域与MRE11相互作用,抑制MRE11的酶活性,最终导致HR和DNA损伤修复(DDR)的抑制。这些过程是通过atm介导的ZBP1在S106位点的磷酸化启动的。与这些发现一致,体外和体内模型在ZBP1过表达后都表现出对PARP抑制剂治疗的敏感性增加。此外,在我们的新辅助尼拉帕尼单药治疗研究(NCT05407841)中,高ZBP1表达与高级别浆液性卵巢癌(HGSOC)对PARP抑制的更好反应和延长PFS相关。本研究描述了ZBP1调节HR的新功能,该功能赋予卵巢癌PARP抑制的合成致死性。因此,ZBP1作为潜在的治疗靶点和对PARP抑制剂反应的生物标志物,以及潜在的其他治疗药物,如铂类似物,对有缺陷的HR具有合成致死性。
{"title":"ZBP1 antagonizes MRE11-mediated DNA end resection and confers synthetic lethality to PARP inhibition in ovarian cancer","authors":"Shen-nan Shi ,&nbsp;Qiuyang Xu ,&nbsp;Zhiqi Liao ,&nbsp;Wenjian Gong ,&nbsp;Yilin Cui ,&nbsp;Jiahao Liu ,&nbsp;Xiaofei Jiao ,&nbsp;Yijie Wu ,&nbsp;Mengshi Luo ,&nbsp;Yuewen Zhang ,&nbsp;Linghui Wang ,&nbsp;Yuanjia Wen ,&nbsp;Wen Pan ,&nbsp;Xuejiao Zhao ,&nbsp;Marilyne Labrie ,&nbsp;Zhiyong Ding ,&nbsp;Gordon B. Mills ,&nbsp;Ding Ma ,&nbsp;Guang-Nian Zhao ,&nbsp;Qinglei Gao ,&nbsp;Yong Fang","doi":"10.1016/j.drup.2025.101319","DOIUrl":"10.1016/j.drup.2025.101319","url":null,"abstract":"<div><div>ZBP1, a classic pattern recognition receptor (PRR), has been implicated in regulating programmed cell death and the innate immune response. However, the role of ZBP1 in the nucleus remains largely undefined. Here, we found that nuclear ZBP1 localizes to the site of DNA double-stranded breaks (DSBs) following DNA damage and impairs homologous recombination (HR) repair through its interaction with MRE11. ZBP1 interacts with MRE11 through RHIM A and B domains and inhibits the enzymatic activity of MRE11, ultimately leading to the suppression of HR and DNA damage repair (DDR). These processes are initiated via ATM-mediated ZBP1 phosphorylation at S106. Consistent with these findings, <em>in vitro</em> and <em>in vivo</em> models both exhibit increased sensitivity to PARP inhibitor treatment following ZBP1 overexpression. Furthermore, in our neoadjuvant niraparib monotherapy study (NCT05407841) higher ZBP1 expression correlates with better response to PARP inhibition and prolonged PFS in high-grade serous ovarian cancer (HGSOC). This study describes a novel function of ZBP1 for regulating HR, which confers synthetic lethality to PARP inhibition in ovarian cancer. ZBP1 thus serves as a potential therapy target and biomarker of response to PARP inhibitors and potentially other therapeutic agents such as platin analogs that are synthetically lethal with defective HR.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"84 ","pages":"Article 101319"},"PeriodicalIF":21.7,"publicationDate":"2025-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145382804","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}
引用次数: 0
Metabolic basis of fatty acid oxidation and immunotherapy resistance with clinical perspectives 脂肪酸氧化代谢基础与免疫治疗耐药的临床研究
IF 21.7 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-10-22 DOI: 10.1016/j.drup.2025.101317
Junfeng Zhang, Jianyou Gu
{"title":"Metabolic basis of fatty acid oxidation and immunotherapy resistance with clinical perspectives","authors":"Junfeng Zhang,&nbsp;Jianyou Gu","doi":"10.1016/j.drup.2025.101317","DOIUrl":"10.1016/j.drup.2025.101317","url":null,"abstract":"","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"84 ","pages":"Article 101317"},"PeriodicalIF":21.7,"publicationDate":"2025-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145363516","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}
引用次数: 0
The novel strategy to overcome the drug-resistant lung cancer: Dual targeting delivery of PROTAC to inhibit cancer-associated fibroblasts and lung cancer cells 克服耐药肺癌的新策略:双靶向递送PROTAC以抑制癌症相关成纤维细胞和肺癌细胞
IF 21.7 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-10-17 DOI: 10.1016/j.drup.2025.101316
Lingmin Zhang , Linlong He , Yinshan Lin , Juyan Wei , Shiqi Tang , Xueping Lei , Xufeng Lin , Dazhi Zhou , Liwu Fu , Yuehua Li , Juyun He , Lu Liang , Xi-Yong Yu
Current pharmacotherapy on the fatal lung cancer is often limited by the development of drug resistance, which significantly contributes to treatment failure. The drug resistance in cancer is associated with tumor microenvironment (TME), particularly with cancer-associated fibroblasts (CAFs). However, the present approaches show little progress in the eliminating lung cancer cells and reversing the TME synergistically. The emergence of nanomedicine offers promising strategies to overcome this challenge. In this study, we developed a proteolysis-targeting chimeras (PROTAC)-based nanodrug, designed to eliminate both lung cancer cells and CAFs, thereby amplifying the therapeutic effects. This nanodrug was constructed by loading dBET6 with US Food and Drug Administration (FDA) approved polymer Poly(lactic-co-glycolic acid) (PLGA), and further camouflaged with the hybrid membranes derived from platelet and lung cancer cells (PLMPD). PLMPD demonstrated excellent dual-targeting capabilities to both lung cancer cells and CAFs, leading to significant apoptosis in both cell types in vitro. We also found that PLMPD could inhibited the growth of Osimertinib-resistant cells. In vivo studies revealed that PLMPD enhanced tumor targeting, effectively inhibited tumor growth, and reversed the tumor-promoting TME in the lung cancer xenograft models. These findings underscore the potential of PLMPD as a promising PROTAC-based nanodrug for lung cancer therapy, offering a new avenue for overcoming drug resistance and improving treatment outcomes.
目前对致命肺癌的药物治疗往往受到耐药性的限制,这是导致治疗失败的重要原因。肿瘤耐药与肿瘤微环境(tumor microenvironment, TME)有关,特别是与癌症相关成纤维细胞(cancer-associated fibroblasts, CAFs)有关。然而,目前的方法在消除肺癌细胞和协同逆转TME方面进展甚微。纳米医学的出现为克服这一挑战提供了有希望的策略。在这项研究中,我们开发了一种基于蛋白水解靶向嵌合体(PROTAC)的纳米药物,旨在消除肺癌细胞和CAFs,从而扩大治疗效果。该纳米药物由美国食品和药物管理局(FDA)批准的聚合物聚乳酸-羟基乙酸(PLGA)负载dBET6构建,并进一步用来自血小板和肺癌细胞的杂交膜(PLMPD)进行伪装。在体外实验中,PLMPD对肺癌细胞和caf均表现出出色的双靶向能力,导致两种细胞类型的显著凋亡。我们还发现PLMPD可以抑制奥西替尼耐药细胞的生长。体内研究显示,PLMPD在肺癌异种移植模型中增强肿瘤靶向性,有效抑制肿瘤生长,逆转促瘤TME。这些发现强调了PLMPD作为一种有前景的基于protac的肺癌治疗纳米药物的潜力,为克服耐药性和改善治疗结果提供了新的途径。
{"title":"The novel strategy to overcome the drug-resistant lung cancer: Dual targeting delivery of PROTAC to inhibit cancer-associated fibroblasts and lung cancer cells","authors":"Lingmin Zhang ,&nbsp;Linlong He ,&nbsp;Yinshan Lin ,&nbsp;Juyan Wei ,&nbsp;Shiqi Tang ,&nbsp;Xueping Lei ,&nbsp;Xufeng Lin ,&nbsp;Dazhi Zhou ,&nbsp;Liwu Fu ,&nbsp;Yuehua Li ,&nbsp;Juyun He ,&nbsp;Lu Liang ,&nbsp;Xi-Yong Yu","doi":"10.1016/j.drup.2025.101316","DOIUrl":"10.1016/j.drup.2025.101316","url":null,"abstract":"<div><div>Current pharmacotherapy on the fatal lung cancer is often limited by the development of drug resistance, which significantly contributes to treatment failure. The drug resistance in cancer is associated with tumor microenvironment (TME), particularly with cancer-associated fibroblasts (CAFs). However, the present approaches show little progress in the eliminating lung cancer cells and reversing the TME synergistically. The emergence of nanomedicine offers promising strategies to overcome this challenge. In this study, we developed a proteolysis-targeting chimeras (PROTAC)-based nanodrug, designed to eliminate both lung cancer cells and CAFs, thereby amplifying the therapeutic effects. This nanodrug was constructed by loading dBET6 with US Food and Drug Administration (FDA) approved polymer Poly(lactic-co-glycolic acid) (PLGA), and further camouflaged with the hybrid membranes derived from platelet and lung cancer cells (PLMPD). PLMPD demonstrated excellent dual-targeting capabilities to both lung cancer cells and CAFs, leading to significant apoptosis in both cell types in vitro. We also found that PLMPD could inhibited the growth of Osimertinib-resistant cells. In vivo studies revealed that PLMPD enhanced tumor targeting, effectively inhibited tumor growth, and reversed the tumor-promoting TME in the lung cancer xenograft models. These findings underscore the potential of PLMPD as a promising PROTAC-based nanodrug for lung cancer therapy, offering a new avenue for overcoming drug resistance and improving treatment outcomes.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"84 ","pages":"Article 101316"},"PeriodicalIF":21.7,"publicationDate":"2025-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145363515","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}
引用次数: 0
期刊
Drug Resistance Updates
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1