首页 > 最新文献

ACS Pharmacology and Translational Science最新文献

英文 中文
Development of Novel 18F-Labeled Selective Orexin-2 Receptor Radioligands for Positron Emission Tomography 用于正电子发射断层扫描的新型18f标记的选择性Orexin-2受体放射配体的开发
IF 3.7 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-10-11 DOI: 10.1021/acsptsci.5c00474
Jian Rong, , , Chunyu Zhao, , , Ahmad F. Chaudhary, , , Jiahui Chen, , , Yinlong Li, , , Xin Zhou, , , Zhendong Song, , , Zhenkun Sun, , , Yabiao Gao, , , Siyan Feng, , , Taoqian Zhao, , , Qi-Long Hu, , , Chongjiao Li, , , Jimmy Patel, , , Hongjie Yuan, , , Achi Haider, , and , Steven H. Liang*, 

The orexin-2 receptor (OX2R), a G protein-coupled receptor activated by the neuropeptides, orexin A and B, plays an integral role in orchestrating motivation, feeding behavior, and the sleep-wake cycle. Pharmacological modulation of OX2R has shown therapeutic potential for a variety of central nervous system (CNS) diseases, most notably narcolepsy and insomnia. Noninvasive imaging of OX2R could enable the visualization of its regional distribution, facilitate assessments of target engagement, and support the development of OX2R-directed therapies. Nonetheless, there are currently no suitable radioligands available for imaging OX2R with positron emission tomography (PET). Herein, we report the design and evaluation of two novel PET ligand candidates, [18F]1 ([18F]OX2-2303) and [18F]2 ([18F]OX2-2304), as potential imaging probes for OX2R. Both candidates exhibit excellent OX2R binding affinity (Ki = 0.1 and 1 nM, respectively) and remarkable selectivity over OX1R (>600-fold). In vitro autoradiography confirmed robust and selective binding to OX2R in rat brain sections. In vivo PET imaging revealed low brain uptake at baseline, attributed to active efflux by P-glycoprotein (P-gp) and/or breast cancer resistance protein (BCRP). Furthermore, pharmacological inhibition of these efflux transporters markedly enhanced brain penetration and OX2R antagonists demonstrated notable blocking effects to OX2R tracers during these conditions. Collectively, [18F]1 ([18F]OX2-2303) and [18F]2 ([18F]OX2-2304) constitute promising chemical starting points for the development of OX2R PET radioligands, although further medicinal chemistry optimization will be required to overcome transporter-mediated efflux from the brain.

食欲素-2受体(OX2R)是一种由神经肽,食欲素a和B激活的G蛋白偶联受体,在协调动机,摄食行为和睡眠-觉醒周期中起着不可或缺的作用。药理调节OX2R已显示出治疗多种中枢神经系统(CNS)疾病的潜力,最显著的是嗜睡症和失眠。OX2R的无创成像可以实现其区域分布的可视化,促进目标参与评估,并支持OX2R定向治疗的发展。尽管如此,目前还没有合适的放射性配体用于正电子发射断层扫描(PET) OX2R成像。在此,我们报道了两种新型PET配体候选体[18F]1 ([18F]OX2-2303)和[18F]2 ([18F]OX2-2304)的设计和评估,作为OX2R的潜在成像探针。这两种候选物都具有出色的OX2R结合亲和力(Ki分别为0.1 nM和1 nM)和对OX1R的显著选择性(>;600倍)。体外放射自显像证实了在大鼠脑切片中与OX2R的强大和选择性结合。体内PET成像显示基线时脑摄取低,归因于p -糖蛋白(P-gp)和/或乳腺癌抵抗蛋白(BCRP)的主动外排。此外,在这些条件下,这些外排转运体的药理抑制显著增强了脑渗透,OX2R拮抗剂对OX2R示踪剂表现出显著的阻断作用。总的来说,[18F]1 ([18F]OX2-2303)和[18F]2 ([18F]OX2-2304)构成了开发OX2R PET放射配体的有希望的化学起点,尽管需要进一步的药物化学优化来克服转运体介导的脑外排。
{"title":"Development of Novel 18F-Labeled Selective Orexin-2 Receptor Radioligands for Positron Emission Tomography","authors":"Jian Rong,&nbsp;, ,&nbsp;Chunyu Zhao,&nbsp;, ,&nbsp;Ahmad F. Chaudhary,&nbsp;, ,&nbsp;Jiahui Chen,&nbsp;, ,&nbsp;Yinlong Li,&nbsp;, ,&nbsp;Xin Zhou,&nbsp;, ,&nbsp;Zhendong Song,&nbsp;, ,&nbsp;Zhenkun Sun,&nbsp;, ,&nbsp;Yabiao Gao,&nbsp;, ,&nbsp;Siyan Feng,&nbsp;, ,&nbsp;Taoqian Zhao,&nbsp;, ,&nbsp;Qi-Long Hu,&nbsp;, ,&nbsp;Chongjiao Li,&nbsp;, ,&nbsp;Jimmy Patel,&nbsp;, ,&nbsp;Hongjie Yuan,&nbsp;, ,&nbsp;Achi Haider,&nbsp;, and ,&nbsp;Steven H. Liang*,&nbsp;","doi":"10.1021/acsptsci.5c00474","DOIUrl":"https://doi.org/10.1021/acsptsci.5c00474","url":null,"abstract":"<p >The orexin-2 receptor (OX<sub>2</sub>R), a G protein-coupled receptor activated by the neuropeptides, orexin A and B, plays an integral role in orchestrating motivation, feeding behavior, and the sleep-wake cycle. Pharmacological modulation of OX<sub>2</sub>R has shown therapeutic potential for a variety of central nervous system (CNS) diseases, most notably narcolepsy and insomnia. Noninvasive imaging of OX<sub>2</sub>R could enable the visualization of its regional distribution, facilitate assessments of target engagement, and support the development of OX<sub>2</sub>R-directed therapies. Nonetheless, there are currently no suitable radioligands available for imaging OX<sub>2</sub>R with positron emission tomography (PET). Herein, we report the design and evaluation of two novel PET ligand candidates, [<sup>18</sup>F]<b>1</b> ([<sup>18</sup>F]OX<sub>2</sub>-2303) and [<sup>18</sup>F]<b>2</b> ([<sup>18</sup>F]OX<sub>2</sub>-2304), as potential imaging probes for OX<sub>2</sub>R. Both candidates exhibit excellent OX<sub>2</sub>R binding affinity (<i>K</i><sub>i</sub> = 0.1 and 1 nM, respectively) and remarkable selectivity over OX<sub>1</sub>R (&gt;600-fold). <i>In vitro</i> autoradiography confirmed robust and selective binding to OX<sub>2</sub>R in rat brain sections. <i>In vivo</i> PET imaging revealed low brain uptake at baseline, attributed to active efflux by P-glycoprotein (P-gp) and/or breast cancer resistance protein (BCRP). Furthermore, pharmacological inhibition of these efflux transporters markedly enhanced brain penetration and OX<sub>2</sub>R antagonists demonstrated notable blocking effects to OX<sub>2</sub>R tracers during these conditions. Collectively, [<sup>18</sup>F]<b>1</b> ([<sup>18</sup>F]OX<sub>2</sub>-2303) and [<sup>18</sup>F]<b>2</b> ([<sup>18</sup>F]OX<sub><b>2</b></sub><b>-</b>2304) constitute promising chemical starting points for the development of OX<sub>2</sub>R PET radioligands, although further medicinal chemistry optimization will be required to overcome transporter-mediated efflux from the brain.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"8 11","pages":"4070–4079"},"PeriodicalIF":3.7,"publicationDate":"2025-10-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/pdf/10.1021/acsptsci.5c00474","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145499724","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Evaluation of the Activity of Monensin and Its Analogs for Modulation of Stem-like Cell Functionality in 2D and 3D Breast Cancer Models 评价莫能菌素及其类似物在二维和三维乳腺癌模型中对干细胞功能调节的活性
IF 3.7 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-10-10 DOI: 10.1021/acsptsci.5c00296
Alicja Urbaniak*, , , Billie Heflin, , , Eric R. Siegel, , , Drew Seale, , , Megan R. Reed, , , James S. Nix, , , Eric U. Yee, , , Marta Jędrzejczyk, , , Greta Klejborowska, , , Natalia Stępczyńska, , , Adam Huczyński, , , Bolni M. Nagalo, , , Timothy C. Chambers, , , Steven R. Post, , , Robert L. Eoff, , , Melanie C. MacNicol, , , Amit K. Tiwari, , , Thomas Kelly, , , Alan J. Tackett, , and , Angus M. MacNicol, 

Monensin (MON) is a polyether ionophore antibiotic of natural origin and is an FDA-approved drug for veterinary use. Recent studies have highlighted its potential anticancer activity in various in vitro and in vivo models. In this study, we evaluated the anti-breast cancer activity of MON and 37 synthetic analog compounds using cell monolayer and organoid models. Through a mini-ring cell viability assay, several compounds were identified that were more potent and selective against breast cancer cells compared to non-cancerous cells, surpassing the activity of parent MON. MON and these compounds induced significant DNA fragmentation, reduced cell migration, and downregulated SOX2 expression. Furthermore, MON and the most potent analog, compound 12, reduced the percentage of CD44+/CD24–/low stem-like cells and diminished colony formation properties. Proteomics analyses revealed that several pathways, including extracellular matrix organization, were significantly dysregulated by MON and compound 12 in breast cancer cells. Among these, TIMP2, a protein associated with the suppression of tumor growth and metastasis, was identified as one of the most prominently upregulated proteins by MON and compound 12 in MDA-MB-231 cells. This finding was also validated in other breast cancer and melanoma cell lines. To simulate breast cancer metastasis to the brain, a human hybrid organoid system: tumor in brain organoid (HOSTBO) model was developed. MON and compound 12 significantly reduced Ki-67 expression within the HOSTBOs, and compound 12 significantly downregulated SOX2 expression. Collectively, MON and compound 12 significantly reduced the proliferation of breast cancer stem-like cells in the organoid models, inhibited their migration, and dysregulated markers associated with stemness, demonstrating their potential as anti-metastatic agents and warranting further clinical development.

莫能菌素(MON)是一种天然聚醚离子载体抗生素,是fda批准的兽药。近年来的研究表明,其潜在的抗癌活性在各种体外和体内模型。在这项研究中,我们利用细胞单层和类器官模型评估了MON和37种合成类似物的抗乳腺癌活性。通过一项微环细胞活力测定,研究人员发现,与非癌细胞相比,几种化合物对乳腺癌细胞具有更强的效力和选择性,超过了亲本MON的活性。MON和这些化合物诱导了显著的DNA断裂,减少了细胞迁移,并下调了SOX2的表达。此外,MON和最有效的类似物化合物12降低了CD44+/CD24 - /低干细胞样细胞的百分比,并降低了集落形成特性。蛋白质组学分析显示,乳腺癌细胞中的一些通路,包括细胞外基质组织,被MON和化合物12显著失调。其中,与抑制肿瘤生长和转移相关的蛋白TIMP2被MON和化合物12鉴定为MDA-MB-231细胞中最显著的上调蛋白之一。这一发现在其他乳腺癌和黑色素瘤细胞系中也得到了证实。为了模拟乳腺癌脑转移,建立了人混合类器官系统:肿瘤在脑类器官(HOSTBO)模型。MON和化合物12显著降低了宿主细胞中Ki-67的表达,化合物12显著下调了SOX2的表达。总的来说,MON和化合物12显著降低了类器官模型中乳腺癌干细胞样细胞的增殖,抑制了它们的迁移,并失调了与干细胞相关的标记物,证明了它们作为抗转移药物的潜力,值得进一步的临床开发。
{"title":"Evaluation of the Activity of Monensin and Its Analogs for Modulation of Stem-like Cell Functionality in 2D and 3D Breast Cancer Models","authors":"Alicja Urbaniak*,&nbsp;, ,&nbsp;Billie Heflin,&nbsp;, ,&nbsp;Eric R. Siegel,&nbsp;, ,&nbsp;Drew Seale,&nbsp;, ,&nbsp;Megan R. Reed,&nbsp;, ,&nbsp;James S. Nix,&nbsp;, ,&nbsp;Eric U. Yee,&nbsp;, ,&nbsp;Marta Jędrzejczyk,&nbsp;, ,&nbsp;Greta Klejborowska,&nbsp;, ,&nbsp;Natalia Stępczyńska,&nbsp;, ,&nbsp;Adam Huczyński,&nbsp;, ,&nbsp;Bolni M. Nagalo,&nbsp;, ,&nbsp;Timothy C. Chambers,&nbsp;, ,&nbsp;Steven R. Post,&nbsp;, ,&nbsp;Robert L. Eoff,&nbsp;, ,&nbsp;Melanie C. MacNicol,&nbsp;, ,&nbsp;Amit K. Tiwari,&nbsp;, ,&nbsp;Thomas Kelly,&nbsp;, ,&nbsp;Alan J. Tackett,&nbsp;, and ,&nbsp;Angus M. MacNicol,&nbsp;","doi":"10.1021/acsptsci.5c00296","DOIUrl":"https://doi.org/10.1021/acsptsci.5c00296","url":null,"abstract":"<p >Monensin (<b>MON</b>) is a polyether ionophore antibiotic of natural origin and is an FDA-approved drug for veterinary use. Recent studies have highlighted its potential anticancer activity in various <i>in vitro</i> and <i>in vivo</i> models. In this study, we evaluated the anti-breast cancer activity of <b>MON</b> and 37 synthetic analog compounds using cell monolayer and organoid models. Through a mini-ring cell viability assay, several compounds were identified that were more potent and selective against breast cancer cells compared to non-cancerous cells, surpassing the activity of parent <b>MON</b>. <b>MON</b> and these compounds induced significant DNA fragmentation, reduced cell migration, and downregulated SOX2 expression. Furthermore, <b>MON</b> and the most potent analog, compound <b>12</b>, reduced the percentage of CD44<sup>+</sup>/CD24<sup>–/low</sup> stem-like cells and diminished colony formation properties. Proteomics analyses revealed that several pathways, including extracellular matrix organization, were significantly dysregulated by <b>MON</b> and compound <b>12</b> in breast cancer cells. Among these, TIMP2, a protein associated with the suppression of tumor growth and metastasis, was identified as one of the most prominently upregulated proteins by <b>MON</b> and compound <b>12</b> in MDA-MB-231 cells. This finding was also validated in other breast cancer and melanoma cell lines. To simulate breast cancer metastasis to the brain, a human hybrid organoid system: tumor in brain organoid (HOSTBO) model was developed. <b>MON</b> and compound <b>12</b> significantly reduced Ki-67 expression within the HOSTBOs, and compound <b>12</b> significantly downregulated SOX2 expression. Collectively, <b>MON</b> and compound <b>12</b> significantly reduced the proliferation of breast cancer stem-like cells in the organoid models, inhibited their migration, and dysregulated markers associated with stemness, demonstrating their potential as anti-metastatic agents and warranting further clinical development.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"8 11","pages":"3965–3983"},"PeriodicalIF":3.7,"publicationDate":"2025-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145499703","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Treatment of Parkinson’s Disease with an Anti-Inflammasome NLRP3 Inhibitor Derived from a Natural Product 从天然产物中提取的抗炎体NLRP3抑制剂治疗帕金森病
IF 3.7 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-10-10 DOI: 10.1021/acsptsci.5c00109
Wenqian Li, , , Jiuliang Zhang, , , Qiang Chen, , , Biaobiao Luo, , , Bo Zhou, , , Jun Wang, , , Yang Yang, , , Yi Liu, , , Shuixiu Wen, , , De-Xin Kong, , , Jian Chen, , , Shaozhong Wei, , , Pierre Duez, , , Avez Sharipov, , , Yan Li, , and , Xuebo Hu*, 

Parkinson’s disease (PD), the second most prevalent neurodegenerative disorder afflicting human health, is primarily characterized by the degeneration of dopaminergic neurons in the midbrain, leading to movement disorders as the main clinical manifestation. Extensive research has demonstrated that the NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome and its accompanying neuroinflammation play a pivotal role in the progression of PD. ST-4, namely 15-oxosteviol, an analogue of the diterpene oridonin, exhibits potent and specific inhibition of NLRP3 in in vitro experiments. The anti-inflammatory effects of ST-4 were evaluated in mouse models of chronic and progressive disorders, in which it showed significant efficacy in ameliorating obesity, type 2 diabetes, and peritonitis. In this study, the potential interest of ST-4 for the treatment of neuroinflammatory diseases was further investigated in a PD mouse model. ST-4 effectively suppressed the activation of the NLRP3 inflammasome induced by lipopolysaccharide in neuronal cells. Additionally, treatment with ST-4 significantly improved various aspects of PD pathology, including behavioral impairments, loss of dopaminergic neurons, alterations in cerebral neurophysiology, and dysregulated gene expression associated with metabolic dysfunction, highlighting its therapeutic potential for the treatment of Parkinson’s disease.

帕金森病(PD)是危害人类健康的第二大常见神经退行性疾病,其主要特征是中脑多巴胺能神经元变性,以运动障碍为主要临床表现。大量研究表明,NOD-、LRR-和pyrin结构域蛋白3 (NLRP3)炎症小体及其伴随的神经炎症在PD的进展中起关键作用。ST-4,即15-oxosteviol,是二萜oriidonin的类似物,在体外实验中显示出对NLRP3的有效和特异性抑制。在慢性和进行性疾病的小鼠模型中,ST-4的抗炎作用被评估,在改善肥胖、2型糖尿病和腹膜炎方面显示出显著的疗效。在本研究中,我们在PD小鼠模型中进一步研究了ST-4治疗神经炎性疾病的潜在兴趣。ST-4能有效抑制脂多糖诱导的神经元细胞NLRP3炎性体的活化。此外,ST-4治疗显著改善了PD病理的各个方面,包括行为障碍、多巴胺能神经元的丧失、脑神经生理学的改变以及与代谢功能障碍相关的基因表达失调,突出了其治疗帕金森病的潜力。
{"title":"Treatment of Parkinson’s Disease with an Anti-Inflammasome NLRP3 Inhibitor Derived from a Natural Product","authors":"Wenqian Li,&nbsp;, ,&nbsp;Jiuliang Zhang,&nbsp;, ,&nbsp;Qiang Chen,&nbsp;, ,&nbsp;Biaobiao Luo,&nbsp;, ,&nbsp;Bo Zhou,&nbsp;, ,&nbsp;Jun Wang,&nbsp;, ,&nbsp;Yang Yang,&nbsp;, ,&nbsp;Yi Liu,&nbsp;, ,&nbsp;Shuixiu Wen,&nbsp;, ,&nbsp;De-Xin Kong,&nbsp;, ,&nbsp;Jian Chen,&nbsp;, ,&nbsp;Shaozhong Wei,&nbsp;, ,&nbsp;Pierre Duez,&nbsp;, ,&nbsp;Avez Sharipov,&nbsp;, ,&nbsp;Yan Li,&nbsp;, and ,&nbsp;Xuebo Hu*,&nbsp;","doi":"10.1021/acsptsci.5c00109","DOIUrl":"https://doi.org/10.1021/acsptsci.5c00109","url":null,"abstract":"<p >Parkinson’s disease (PD), the second most prevalent neurodegenerative disorder afflicting human health, is primarily characterized by the degeneration of dopaminergic neurons in the midbrain, leading to movement disorders as the main clinical manifestation. Extensive research has demonstrated that the NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome and its accompanying neuroinflammation play a pivotal role in the progression of PD. ST-4, namely 15-oxosteviol, an analogue of the diterpene oridonin, exhibits potent and specific inhibition of NLRP3 in <i>in vitro</i> experiments. The anti-inflammatory effects of ST-4 were evaluated in mouse models of chronic and progressive disorders, in which it showed significant efficacy in ameliorating obesity, type 2 diabetes, and peritonitis. In this study, the potential interest of ST-4 for the treatment of neuroinflammatory diseases was further investigated in a PD mouse model. ST-4 effectively suppressed the activation of the NLRP3 inflammasome induced by lipopolysaccharide in neuronal cells. Additionally, treatment with ST-4 significantly improved various aspects of PD pathology, including behavioral impairments, loss of dopaminergic neurons, alterations in cerebral neurophysiology, and dysregulated gene expression associated with metabolic dysfunction, highlighting its therapeutic potential for the treatment of Parkinson’s disease.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"8 11","pages":"3896–3909"},"PeriodicalIF":3.7,"publicationDate":"2025-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145499705","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Mapping the Progression of Therapy-Induced Senescence to Therapy Tolerance: An Evolutionarily Conserved Mechanism for Optimizing Cancer Treatment with Senotherapeutics 从治疗诱导的衰老到治疗耐受:一种进化保守的机制来优化老年治疗药物对癌症的治疗
IF 3.7 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-10-10 DOI: 10.1021/acsptsci.5c00374
Gargi Mukherjee, , , Neha Dutta, , , Muthumeena Ramanathan, , , Kankonika Bhattacharyya, , , Alabhya Das, , , Shaista Haider, , , Goutam Chowdhury, , and , Anindita Chakrabarty*, 

Therapy-induced senescence (TIS) is a reversible growth arrest induced by anticancer treatments, which may contribute to the development of long-term therapy resistance in tumor cells. Senotherapeutics, agents targeting senescent cells, are being tested in clinical trials to improve patient outcomes. Due to the transient nature of TIS, we hypothesized that senolytics would be most effective when administered at the appropriate time. We created a reliable TIS cell line model in triple-negative breast cancer (TNBC) using experimental drug YM155. We observed that a single dose of YM155 triggers a brief senescence, leading to a persistent drug-tolerant state that cannot be reversed by redosing. This reversibility is not limited to cancer cells. It extends to noncancerous human cells and live zebrafish larvae, suggesting a rapid adaptation mechanism against xenobiotics. We identified transforming growth factor-β (TGF-β), a cytokine linked to TNBC chemoresistance, as being expressed alongside the emergence of drug tolerance. We inhibited TGF-β signaling to eliminate the tolerant phenotype and promote the clearance of cancer cells by immune cells. However, this was most effective within a specific time window after TIS induction. We suggest that the timely use of senotherapeutics could improve the effectiveness of anticancer drugs in clinical settings.

治疗性衰老(TIS)是一种由抗癌治疗引起的可逆性生长停滞,可能有助于肿瘤细胞长期耐药的发展。针对衰老细胞的衰老疗法正在临床试验中进行测试,以改善患者的治疗效果。由于TIS的短暂性,我们假设在适当的时间使用抗衰老药物是最有效的。我们使用实验药物YM155在三阴性乳腺癌(TNBC)中建立了可靠的TIS细胞系模型。我们观察到单剂量的YM155触发短暂的衰老,导致持续的耐药状态,不能通过重新给药来逆转。这种可逆性并不局限于癌细胞。它延伸到非癌细胞和活斑马鱼幼虫,表明一种针对外源药物的快速适应机制。我们确定了转化生长因子-β (TGF-β),一种与TNBC化疗耐药相关的细胞因子,随着药物耐受性的出现而表达。我们通过抑制TGF-β信号通路消除耐药表型,促进免疫细胞对癌细胞的清除。然而,这种方法在TIS诱导后的特定时间窗内最为有效。我们认为,及时使用老年治疗药物可以提高临床抗癌药物的有效性。
{"title":"Mapping the Progression of Therapy-Induced Senescence to Therapy Tolerance: An Evolutionarily Conserved Mechanism for Optimizing Cancer Treatment with Senotherapeutics","authors":"Gargi Mukherjee,&nbsp;, ,&nbsp;Neha Dutta,&nbsp;, ,&nbsp;Muthumeena Ramanathan,&nbsp;, ,&nbsp;Kankonika Bhattacharyya,&nbsp;, ,&nbsp;Alabhya Das,&nbsp;, ,&nbsp;Shaista Haider,&nbsp;, ,&nbsp;Goutam Chowdhury,&nbsp;, and ,&nbsp;Anindita Chakrabarty*,&nbsp;","doi":"10.1021/acsptsci.5c00374","DOIUrl":"https://doi.org/10.1021/acsptsci.5c00374","url":null,"abstract":"<p >Therapy-induced senescence (TIS) is a reversible growth arrest induced by anticancer treatments, which may contribute to the development of long-term therapy resistance in tumor cells. Senotherapeutics, agents targeting senescent cells, are being tested in clinical trials to improve patient outcomes. Due to the transient nature of TIS, we hypothesized that senolytics would be most effective when administered at the appropriate time. We created a reliable TIS cell line model in triple-negative breast cancer (TNBC) using experimental drug YM155. We observed that a single dose of YM155 triggers a brief senescence, leading to a persistent drug-tolerant state that cannot be reversed by redosing. This reversibility is not limited to cancer cells. It extends to noncancerous human cells and live zebrafish larvae, suggesting a rapid adaptation mechanism against xenobiotics. We identified transforming growth factor-β (TGF-β), a cytokine linked to TNBC chemoresistance, as being expressed alongside the emergence of drug tolerance. We inhibited TGF-β signaling to eliminate the tolerant phenotype and promote the clearance of cancer cells by immune cells. However, this was most effective within a specific time window after TIS induction. We suggest that the timely use of senotherapeutics could improve the effectiveness of anticancer drugs in clinical settings.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"8 11","pages":"4001–4016"},"PeriodicalIF":3.7,"publicationDate":"2025-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145499704","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Crescent-Shaped Xanthone–Benzimidazole Conjugates as New Target Probes for Telomeric G-Quadruplex DNA and Specific Cytotoxicity on Cancer Cells 新月形杂蒽酮-苯并咪唑缀合物作为端粒g -四重体DNA的新靶探针及其对癌细胞的特异性细胞毒性
IF 3.7 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-10-09 DOI: 10.1021/acsptsci.5c00408
Pulakesh Pramanik, , , Bappa Maiti, , and , Santanu Bhattacharya*, 

Xanthone, an oxygenated heterotricyclic phytochemical, has recently emerged as a promising scaffold in cancer research due to its multitargeted anticancer potential. Recent studies demonstrate its affinity for G-quadruplex (G4) DNA structures. In this context, herein, we describe the design and synthesis of a series of xanthone–benzimidazole conjugates to investigate their specific cytotoxic effects on cancer cells through the stabilization of telomeric G4 DNA structures. Comprehensive in vitro biophysical and cellular experiments were performed to screen the most effective compounds for targeting telomeric G4 DNA structures among them. The structure–activity relationship (SAR) of quadruplex-compound interactions was documented as well. The current study reveals that two compounds, XDBHEP and XDBAEP, exhibit maximum binding affinity and selectivity toward the antiparallel telomeric form of G4 DNA. These compounds stabilize this G4 DNA structure through binding within the DNA groove loci and exhibit a 1:1 molar binding stoichiometry. The specific cytotoxic effect of these compounds on different types of cancer cells, including A549, T-47D, and MCF-7, and their apoptotic-mediated cell death was further demonstrated by several in vitro cellular experiments. In addition, blood compatibility, ADME studies, pharmacokinetics, and biodistribution studies were also performed to assess the potential of these compounds for further in vivo and clinical investigations. Based on the current study, the selective antiparallel telomeric G4 DNA targeting properties and specific cancer cell cytotoxicity effects of xanthone–benzimidazole conjugates could provide valuable information to support the research community in the future development of new anticancer drugs through G4 DNA stabilization based on this pharmacophore.

山酮是一种含氧杂三环植物化学物质,由于其多靶点的抗癌潜力,近年来在癌症研究中成为一种很有前途的支架。最近的研究表明它对g -四重体(G4) DNA结构具有亲和力。在此背景下,我们设计和合成了一系列的杂蒽酮-苯并咪唑缀合物,通过稳定端粒G4 DNA结构来研究它们对癌细胞的特异性细胞毒性作用。通过全面的体外生物物理和细胞实验来筛选其中最有效的靶向G4 DNA端粒结构的化合物。并对四复相化合物相互作用的构效关系进行了研究。目前的研究表明,XDBHEP和XDBAEP两种化合物对G4 DNA的反平行端粒形式表现出最大的结合亲和力和选择性。这些化合物通过在DNA凹槽位点内结合来稳定G4 DNA结构,并表现出1:1的摩尔结合化学计量。这些化合物对不同类型的癌细胞(包括A549、T-47D和MCF-7)的特异性细胞毒作用及其凋亡介导的细胞死亡在几个体外细胞实验中得到了进一步证实。此外,还进行了血液相容性、ADME研究、药代动力学和生物分布研究,以评估这些化合物在进一步体内和临床研究中的潜力。基于目前的研究,杂蒽酮-苯并咪唑偶联物的选择性反平行端粒G4 DNA靶向特性和特异性癌细胞细胞毒性作用可以为未来研究界基于该药效团稳定G4 DNA开发新的抗癌药物提供有价值的信息。
{"title":"Crescent-Shaped Xanthone–Benzimidazole Conjugates as New Target Probes for Telomeric G-Quadruplex DNA and Specific Cytotoxicity on Cancer Cells","authors":"Pulakesh Pramanik,&nbsp;, ,&nbsp;Bappa Maiti,&nbsp;, and ,&nbsp;Santanu Bhattacharya*,&nbsp;","doi":"10.1021/acsptsci.5c00408","DOIUrl":"https://doi.org/10.1021/acsptsci.5c00408","url":null,"abstract":"<p >Xanthone, an oxygenated heterotricyclic phytochemical, has recently emerged as a promising scaffold in cancer research due to its multitargeted anticancer potential. Recent studies demonstrate its affinity for G-quadruplex (G4) DNA structures. In this context, herein, we describe the design and synthesis of a series of xanthone–benzimidazole conjugates to investigate their specific cytotoxic effects on cancer cells through the stabilization of telomeric G4 DNA structures. Comprehensive <i>in vitro</i> biophysical and cellular experiments were performed to screen the most effective compounds for targeting telomeric G4 DNA structures among them. The structure–activity relationship (SAR) of quadruplex-compound interactions was documented as well. The current study reveals that two compounds, XDBHEP and XDBAEP, exhibit maximum binding affinity and selectivity toward the antiparallel telomeric form of G4 DNA. These compounds stabilize this G4 DNA structure through binding within the DNA groove loci and exhibit a 1:1 molar binding stoichiometry. The specific cytotoxic effect of these compounds on different types of cancer cells, including A549, T-47D, and MCF-7, and their apoptotic-mediated cell death was further demonstrated by several <i>in vitro</i> cellular experiments. In addition, blood compatibility, ADME studies, pharmacokinetics, and biodistribution studies were also performed to assess the potential of these compounds for further <i>in vivo</i> and clinical investigations. Based on the current study, the selective antiparallel telomeric G4 DNA targeting properties and specific cancer cell cytotoxicity effects of xanthone–benzimidazole conjugates could provide valuable information to support the research community in the future development of new anticancer drugs through G4 DNA stabilization based on this pharmacophore.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"8 11","pages":"4017–4037"},"PeriodicalIF":3.7,"publicationDate":"2025-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145499717","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Development of a Cell-Based Recombinant Green Fluorescent Protein Assay System for Generalized Discovery of Viral Protease Inhibitors 基于细胞的重组绿色荧光蛋白检测系统的建立,用于广泛发现病毒蛋白酶抑制剂
IF 3.7 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-10-09 DOI: 10.1021/acsptsci.5c00262
Yan Feng, , , Xiaoyan Wu, , , Ruiting Chen, , , Yao Fan, , , Changping Xu, , , Chenjie Fang, , , Huimin Sun, , , Shuling Jian, , , Jiasheng Song, , and , Beibei Wu*, 

Viral proteases are critical targets for antiviral drug development, but current screening methods for protease inhibitors often require high biosafety levels or lack cell-based relevance. Here, we developed a novel cell-based assay system utilizing recombinant green fluorescent protein (GFP) technology, designated as DIFF-recombinant GFP (DIFF-rGFP), for potentially high-throughput screening of viral protease inhibitors. By systematically investigating potential insertion sites within the green fluorescent protein (GFP), we constructed a series of recombinant green fluorescent proteins (rGFPs) that accommodate exogenous protease cleavage sequences. Using the 3-Chymotrypsin like protease (3CLpro) of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) as a model, we demonstrated that the DIFF-rGFP assay relies on the coexpression of rGFP and the protease, with fluorescence intensity increasing upon inhibitor action. This assay eliminates the need for high biosafety laboratories and is performed at the cellular level. For proof of concept, we validated this method using two well-characterized SARS-CoV-2 3CLpro inhibitors, GC376 and ensitrelvir, to demonstrate its applicability for inhibitor screening. Our results indicate that the DIFF-rGFP assay is a safe, efficient, and reliable platform for identifying viral protease inhibitors with potential applications in accelerating antiviral drug discovery.

病毒蛋白酶是抗病毒药物开发的关键靶点,但目前蛋白酶抑制剂的筛选方法通常要求较高的生物安全水平或缺乏基于细胞的相关性。在这里,我们开发了一种新的基于细胞的检测系统,利用重组绿色荧光蛋白(GFP)技术,被命名为diff -重组绿色荧光蛋白(DIFF-rGFP),用于潜在的高通量筛选病毒蛋白酶抑制剂。通过系统地研究绿色荧光蛋白(GFP)的潜在插入位点,我们构建了一系列适应外源蛋白酶裂解序列的重组绿色荧光蛋白(rgfp)。以SARS-CoV-2的3-糜凝胰蛋白酶样蛋白酶(3CLpro)为模型,我们证明了DIFF-rGFP检测依赖于rGFP和蛋白酶的共表达,在抑制剂作用下荧光强度增加。该检测无需高生物安全性实验室,可在细胞水平上进行。为了验证概念,我们使用了两种特征良好的SARS-CoV-2 3CLpro抑制剂GC376和ensitrelvir来验证该方法,以证明其用于抑制剂筛选的适用性。我们的研究结果表明,DIFF-rGFP分析是一个安全、高效、可靠的平台,用于鉴定病毒蛋白酶抑制剂,在加速抗病毒药物的发现方面具有潜在的应用前景。
{"title":"Development of a Cell-Based Recombinant Green Fluorescent Protein Assay System for Generalized Discovery of Viral Protease Inhibitors","authors":"Yan Feng,&nbsp;, ,&nbsp;Xiaoyan Wu,&nbsp;, ,&nbsp;Ruiting Chen,&nbsp;, ,&nbsp;Yao Fan,&nbsp;, ,&nbsp;Changping Xu,&nbsp;, ,&nbsp;Chenjie Fang,&nbsp;, ,&nbsp;Huimin Sun,&nbsp;, ,&nbsp;Shuling Jian,&nbsp;, ,&nbsp;Jiasheng Song,&nbsp;, and ,&nbsp;Beibei Wu*,&nbsp;","doi":"10.1021/acsptsci.5c00262","DOIUrl":"https://doi.org/10.1021/acsptsci.5c00262","url":null,"abstract":"<p >Viral proteases are critical targets for antiviral drug development, but current screening methods for protease inhibitors often require high biosafety levels or lack cell-based relevance. Here, we developed a novel cell-based assay system utilizing recombinant green fluorescent protein (GFP) technology, designated as DIFF-recombinant GFP (DIFF-rGFP), for potentially high-throughput screening of viral protease inhibitors. By systematically investigating potential insertion sites within the green fluorescent protein (GFP), we constructed a series of recombinant green fluorescent proteins (rGFPs) that accommodate exogenous protease cleavage sequences. Using the 3-Chymotrypsin like protease (3CL<sup>pro</sup>) of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) as a model, we demonstrated that the DIFF-rGFP assay relies on the coexpression of rGFP and the protease, with fluorescence intensity increasing upon inhibitor action. This assay eliminates the need for high biosafety laboratories and is performed at the cellular level. For proof of concept, we validated this method using two well-characterized SARS-CoV-2 3CL<sup>pro</sup> inhibitors, GC376 and ensitrelvir, to demonstrate its applicability for inhibitor screening. Our results indicate that the DIFF-rGFP assay is a safe, efficient, and reliable platform for identifying viral protease inhibitors with potential applications in accelerating antiviral drug discovery.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"8 11","pages":"3944–3952"},"PeriodicalIF":3.7,"publicationDate":"2025-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145499667","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cannabidiol in Sports: A Brazilian Perspective 大麻二酚在体育:巴西的观点
IF 3.7 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-10-08 DOI: 10.1021/acsptsci.5c00580
Tatyana Nery, , , Jose Wilson NV Andrade, , , Jimmy Fardin Rocha, , , Ana Paula Pinto de Araújo, , and , Aderbal Silva Aguiar Jr*, 

This Viewpoint examines the emerging role of cannabidiol (CBD) in sports medicine, with a particular emphasis on its potential to support athlete health through indirect mechanisms. Rather than acting as a direct performance enhancer, CBD may contribute to improved readiness for training and competition by promoting better sleep, alleviating anxiety, and accelerating recovery. We also discuss challenges for antidoping compliance, including product contamination and regulatory gaps in Brazil, and highlight CBD’s promise as a safer alternative to opioids for pain management in athletes.

本观点探讨了大麻二酚(CBD)在运动医学中的新兴作用,特别强调其通过间接机制支持运动员健康的潜力。CBD不是作为直接的表现增强剂,而是通过促进更好的睡眠,减轻焦虑和加速恢复,有助于提高训练和比赛的准备程度。我们还讨论了反兴奋剂合规方面的挑战,包括巴西的产品污染和监管缺口,并强调CBD有望成为阿片类药物更安全的替代品,用于运动员的疼痛管理。
{"title":"Cannabidiol in Sports: A Brazilian Perspective","authors":"Tatyana Nery,&nbsp;, ,&nbsp;Jose Wilson NV Andrade,&nbsp;, ,&nbsp;Jimmy Fardin Rocha,&nbsp;, ,&nbsp;Ana Paula Pinto de Araújo,&nbsp;, and ,&nbsp;Aderbal Silva Aguiar Jr*,&nbsp;","doi":"10.1021/acsptsci.5c00580","DOIUrl":"https://doi.org/10.1021/acsptsci.5c00580","url":null,"abstract":"<p >This Viewpoint examines the emerging role of cannabidiol (CBD) in sports medicine, with a particular emphasis on its potential to support athlete health through indirect mechanisms. Rather than acting as a direct performance enhancer, CBD may contribute to improved readiness for training and competition by promoting better sleep, alleviating anxiety, and accelerating recovery. We also discuss challenges for antidoping compliance, including product contamination and regulatory gaps in Brazil, and highlight CBD’s promise as a safer alternative to opioids for pain management in athletes.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"8 11","pages":"4189–4192"},"PeriodicalIF":3.7,"publicationDate":"2025-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/pdf/10.1021/acsptsci.5c00580","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145499666","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Chrono-Pharmacology for Cancer: Harnessing Circadian Regulations of the Cell Cycle and Immune Response Dynamics for Precision Therapy 癌症的时间药理学:利用细胞周期的昼夜节律调节和精确治疗的免疫反应动力学
IF 3.7 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-10-08 DOI: 10.1021/acsptsci.5c00617
Murlidhar Madhukar, , , Sandip Das, , , Kavipriya M, , , Sandipan Ray*, , and , Aravind Kumar Rengan*, 

The circadian rhythms and cell cycle are closely interlinked, creating a fundamental regulatory axis vital for tissue homeostasis, which is frequently dysregulated in cancers. The circadian apparatus, which is regulated by the core clock components (BMAL1, CLOCK, PER, and CRY in mammals), establishes temporal order on cell proliferation by rhythmically regulating important cell cycle regulators such as WEE1, p21, and the oncogene MYC. This is frequently accomplished through overlapped signaling nodes that include particular kinases and ubiquitin ligases (e.g., FBXW7). Mounting evidence implicates disruption of this circadian clock-cell cycle synchrony, arising from genetic or environmental factors, as a significant contributor to tumorigenesis and progression via impacts on DNA repair fidelity, oncogene stability, and tumor suppressor pathways. This review critically evaluates the new concept of chrono-pharmacology for cancer, focusing on the substantial effects and side effects of different anticancer drugs that depend on the time-of-day efficacy. We discussed some interesting examples, like HSP90 inhibitors (ganetespib), HDAC inhibitors (quisinostat), topoisomerase inhibitors (doxorubicin), and BCL-2 family antagonists (Obatoclax, TW-37), whose therapeutic activities are tightly regulated by circadian control over their molecular targets, pharmacokinetic processes, and downstream physiological pathways. Furthermore, the circadian influence extends to the tumor microenvironment and antitumor immunity, suggesting novel chrono-immunotherapy approaches. By putting together the molecular bases of these temporal dynamics, this review underscores the significant potential of chronotherapy─the timed administration of drugs to improve cancer treatment by enhancing therapeutic indices and paving the way for personalized, temporally optimized oncology strategies.

昼夜节律和细胞周期紧密相连,创造了一个对组织稳态至关重要的基本调节轴,而组织稳态在癌症中经常失调。昼夜节律装置由核心时钟组件(哺乳动物中的BMAL1、clock、PER和CRY)调节,通过有节奏地调节重要的细胞周期调节因子,如WEE1、p21和癌基因MYC,建立细胞增殖的时间顺序。这通常是通过包括特定激酶和泛素连接酶(如FBXW7)的重叠信号节点完成的。越来越多的证据表明,由遗传或环境因素引起的生物钟-细胞周期同步的破坏,通过对DNA修复保真度、癌基因稳定性和肿瘤抑制途径的影响,是肿瘤发生和进展的重要因素。这篇综述批判性地评估了癌症时间药理学的新概念,重点关注不同抗癌药物依赖于一天中的时间疗效的实质效应和副作用。我们讨论了一些有趣的例子,如HSP90抑制剂(ganetespib)、HDAC抑制剂(quisinostat)、拓扑异构酶抑制剂(阿霉素)和BCL-2家族拮抗剂(Obatoclax, w -37),它们的治疗活性受到其分子靶点、药代动力学过程和下游生理途径的昼夜节律控制的严格调节。此外,昼夜节律影响延伸到肿瘤微环境和抗肿瘤免疫,提示新的时间免疫治疗方法。通过综合这些时间动态的分子基础,本综述强调了时间疗法的巨大潜力──通过提高治疗指标和为个性化、时间优化的肿瘤策略铺平道路,定时给药来改善癌症治疗。
{"title":"Chrono-Pharmacology for Cancer: Harnessing Circadian Regulations of the Cell Cycle and Immune Response Dynamics for Precision Therapy","authors":"Murlidhar Madhukar,&nbsp;, ,&nbsp;Sandip Das,&nbsp;, ,&nbsp;Kavipriya M,&nbsp;, ,&nbsp;Sandipan Ray*,&nbsp;, and ,&nbsp;Aravind Kumar Rengan*,&nbsp;","doi":"10.1021/acsptsci.5c00617","DOIUrl":"https://doi.org/10.1021/acsptsci.5c00617","url":null,"abstract":"<p >The circadian rhythms and cell cycle are closely interlinked, creating a fundamental regulatory axis vital for tissue homeostasis, which is frequently dysregulated in cancers. The circadian apparatus, which is regulated by the core clock components (BMAL1, CLOCK, PER, and CRY in mammals), establishes temporal order on cell proliferation by rhythmically regulating important cell cycle regulators such as WEE1, p21, and the oncogene MYC. This is frequently accomplished through overlapped signaling nodes that include particular kinases and ubiquitin ligases (e.g., FBXW7). Mounting evidence implicates disruption of this circadian clock-cell cycle synchrony, arising from genetic or environmental factors, as a significant contributor to tumorigenesis and progression via impacts on DNA repair fidelity, oncogene stability, and tumor suppressor pathways. This review critically evaluates the new concept of chrono-pharmacology for cancer, focusing on the substantial effects and side effects of different anticancer drugs that depend on the time-of-day efficacy. We discussed some interesting examples, like HSP90 inhibitors (ganetespib), HDAC inhibitors (quisinostat), topoisomerase inhibitors (doxorubicin), and BCL-2 family antagonists (Obatoclax, TW-37), whose therapeutic activities are tightly regulated by circadian control over their molecular targets, pharmacokinetic processes, and downstream physiological pathways. Furthermore, the circadian influence extends to the tumor microenvironment and antitumor immunity, suggesting novel chrono-immunotherapy approaches. By putting together the molecular bases of these temporal dynamics, this review underscores the significant potential of chronotherapy─the timed administration of drugs to improve cancer treatment by enhancing therapeutic indices and paving the way for personalized, temporally optimized oncology strategies.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"8 11","pages":"3821–3834"},"PeriodicalIF":3.7,"publicationDate":"2025-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145499665","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Conformational Heterogeneity Underlying Divergent Signaling in Class A G Protein-Coupled Receptors A类G蛋白偶联受体分化信号的构象异质性
IF 3.7 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-10-07 DOI: 10.1021/acsptsci.5c00102
Kyriakos Georgiou,  and , Antonios Kolocouris*, 

Class A G protein-coupled receptors (GPCRs) are targets for ∼36% of commercial drugs. GPCRs in their apo-forms exhibit conformational heterogeneity, and more than a single active and inactive conformation exists in equilibrium. Distinct transient conformational states can be significantly populated and can be coupled with different agonists, transducers, and effectors, giving rise to divergent signaling pathways. The characterization of such transient conformational states, which may have eluded identification by X-ray crystallography and cryogenic electron microscopy, can be achieved through a combination of biophysical techniques, such as nuclear magnetic resonance, double electron–electron resonance spectroscopy, single-molecule fluorescence microscopy, molecular dynamics simulations, and mass spectrometry. We review findings about the functional, conformational states of four class A GPCRs, including detailed results for the adenosine A2A and β2 adrenergic receptors and important observations for the β1 and μ opioid receptors. The identification of ligands that can bind to distinct conformations, e.g., agonists that activate favorable pathways while inhibiting deleterious ones, represents an important goal in drug development.

A类G蛋白偶联受体(gpcr)是约36%的商业药物的靶标。在它们的载子形式的gpcr表现出构象异质性,并超过一个单一的活性和非活性构象存在于平衡。不同的瞬态构象状态可以显著填充,并且可以与不同的激动剂、换能器和效应器耦合,从而产生不同的信号通路。这种瞬态构象的表征,可能无法通过x射线晶体学和低温电子显微镜识别,可以通过结合生物物理技术,如核磁共振,双电子-电子共振光谱,单分子荧光显微镜,分子动力学模拟和质谱来实现。本文综述了四种A类gpcr的功能和构象状态,包括腺苷A2A和β2肾上腺素能受体的详细结果以及β1和μ阿片受体的重要观察结果。识别可以结合不同构象的配体,例如,激活有利途径同时抑制有害途径的激动剂,是药物开发的一个重要目标。
{"title":"Conformational Heterogeneity Underlying Divergent Signaling in Class A G Protein-Coupled Receptors","authors":"Kyriakos Georgiou,&nbsp; and ,&nbsp;Antonios Kolocouris*,&nbsp;","doi":"10.1021/acsptsci.5c00102","DOIUrl":"https://doi.org/10.1021/acsptsci.5c00102","url":null,"abstract":"<p >Class A G protein-coupled receptors (GPCRs) are targets for ∼36% of commercial drugs. GPCRs in their apo-forms exhibit conformational heterogeneity, and more than a single active and inactive conformation exists in equilibrium. Distinct transient conformational states can be significantly populated and can be coupled with different agonists, transducers, and effectors, giving rise to divergent signaling pathways. The characterization of such transient conformational states, which may have eluded identification by X-ray crystallography and cryogenic electron microscopy, can be achieved through a combination of biophysical techniques, such as nuclear magnetic resonance, double electron–electron resonance spectroscopy, single-molecule fluorescence microscopy, molecular dynamics simulations, and mass spectrometry. We review findings about the functional, conformational states of four class A GPCRs, including detailed results for the adenosine A<sub>2A</sub> and β<sub>2</sub> adrenergic receptors and important observations for the β<sub>1</sub> and μ opioid receptors. The identification of ligands that can bind to distinct conformations, e.g., agonists that activate favorable pathways while inhibiting deleterious ones, represents an important goal in drug development.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"8 11","pages":"3691–3728"},"PeriodicalIF":3.7,"publicationDate":"2025-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/pdf/10.1021/acsptsci.5c00102","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145499664","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Development and Preclinical Evaluation of a Zirconium-89-Labeled Immunoconjugate for Positron Emission Tomography Imaging of Leukemia Inhibitory Factor in Glioblastoma 锆-89标记免疫偶联物用于胶质瘤白血病抑制因子正电子发射断层成像的研制及临床前评价
IF 3.7 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-10-07 DOI: 10.1021/acsptsci.5c00093
Emily Betancourt Fernández, , , Julia Baguña Torres, , , Raffaella Iurlaro, , , Isabel Cuartas, , , Martha Sahylí Ortega Pijeira, , , Sofía Rodríguez Fernández, , , Sergi Velasco-Vila, , , Joan Seoane*, , and , J. Raul Herance*, 

Leukemia Inhibitory Factor (LIF) is a pleiotropic cytokine secreted by tumor cells to evade immune detection, contributing to tumor progression and resistance to therapy. Targeting LIF has emerged as a promising strategy, with anti-LIF therapies in clinical trials across a variety of cancers. Glioblastoma, a highly aggressive LIF-secreting brain tumor, is a critical target for these emerging therapies. This study aimed to develop an anti-LIF immunoPET agent for monitoring LIF expression in vivo, improving detection and targeted treatment strategies for glioblastoma. An anti-LIF antibody was conjugated to p-SCN-Bz-DFO and radiolabeled with positron-emitting zirconium-89 (89Zr). Target binding properties and stability of the radioimmunoconjugate were assessed by ELISA and size exclusion chromatography. The biodistribution of [89Zr]Zr-DFO-anti-LIF was evaluated by PET/CT imaging in an orthotopic glioblastoma mouse model with LIF-positive (GL261N) and LIF-negative (GL261N-CRISPR/LIF) tumors at 24, 48, and 72 hours post-administration. Tumor LIF levels were measured ex vivo by immunohistochemistry. Mass spectrometry determined 2.4 ± 0.3 chelators per antibody molecule. Competitive ELISA demonstrated unaltered affinity post-conjugation. Radiolabeling at a 1 MBq of 89Zr per 5 μg of anti-LIF ratio achieved >68% yield, >95% purity, and 0.17 ± 0.03 MBq/μg specific activity. The radioimmunoconjugate remained >90% intact after 72 h in both saline and mouse serum. PET imaging revealed specific accumulation in LIF-positive brain tumors in vivo (6 ± 1.26 % ID/mL at 72 h), which was 2-fold higher than that observed in the GL261N-CRISPR/LIF model. [89Zr]Zr-DFO-anti-LIF was successfully synthesized, exhibiting specificity and stability in vitro and in vivo, thus supporting its potential for glioblastoma monitoring, as well as guiding anti-LIF therapies.

白血病抑制因子(Leukemia Inhibitory Factor, LIF)是肿瘤细胞为逃避免疫检测而分泌的一种多效性细胞因子,有助于肿瘤的进展和对治疗的抵抗。靶向LIF已成为一种有前景的策略,抗LIF疗法已在各种癌症的临床试验中得到应用。胶质母细胞瘤是一种高度侵袭性的分泌liff的脑肿瘤,是这些新兴疗法的关键靶点。本研究旨在开发一种抗LIF免疫pet试剂,用于监测体内LIF的表达,提高胶质母细胞瘤的检测和靶向治疗策略。将抗lif抗体偶联到p-SCN-Bz-DFO上,并用正电子发射锆-89 (89Zr)进行放射性标记。采用酶联免疫吸附法和大小排斥层析法评价放射免疫偶联物的靶结合性能和稳定性。[89Zr]Zr-DFO-anti-LIF在给药后24、48和72小时的原位胶质母细胞瘤小鼠模型中,通过PET/CT成像评估LIF阳性(GL261N)和LIF阴性(GL261N- crispr /LIF)肿瘤的生物分布。体外免疫组化法测定肿瘤LIF水平。质谱测定每个抗体分子有2.4±0.3个螯合剂。竞争性酶联免疫吸附试验显示偶联后亲和力不变。以每5 μg抗lif比为1 MBq (89Zr)进行放射性标记,产率为68%,纯度为95%,比活性为0.17±0.03 MBq/μg。72小时后,放射免疫偶联物在生理盐水和小鼠血清中均保持90%完整。PET显像显示,活体中LIF阳性脑肿瘤的特异性积累(72 h时为6±1.26% ID/mL),比GL261N-CRISPR/LIF模型高2倍。[89Zr]Zr-DFO-anti-LIF成功合成,体外和体内均表现出特异性和稳定性,从而支持其在胶质母细胞瘤监测中的潜力,并指导抗lif治疗。
{"title":"Development and Preclinical Evaluation of a Zirconium-89-Labeled Immunoconjugate for Positron Emission Tomography Imaging of Leukemia Inhibitory Factor in Glioblastoma","authors":"Emily Betancourt Fernández,&nbsp;, ,&nbsp;Julia Baguña Torres,&nbsp;, ,&nbsp;Raffaella Iurlaro,&nbsp;, ,&nbsp;Isabel Cuartas,&nbsp;, ,&nbsp;Martha Sahylí Ortega Pijeira,&nbsp;, ,&nbsp;Sofía Rodríguez Fernández,&nbsp;, ,&nbsp;Sergi Velasco-Vila,&nbsp;, ,&nbsp;Joan Seoane*,&nbsp;, and ,&nbsp;J. Raul Herance*,&nbsp;","doi":"10.1021/acsptsci.5c00093","DOIUrl":"https://doi.org/10.1021/acsptsci.5c00093","url":null,"abstract":"<p >Leukemia Inhibitory Factor (LIF) is a pleiotropic cytokine secreted by tumor cells to evade immune detection, contributing to tumor progression and resistance to therapy. Targeting LIF has emerged as a promising strategy, with anti-LIF therapies in clinical trials across a variety of cancers. Glioblastoma, a highly aggressive LIF-secreting brain tumor, is a critical target for these emerging therapies. This study aimed to develop an anti-LIF immunoPET agent for monitoring LIF expression in vivo, improving detection and targeted treatment strategies for glioblastoma. An anti-LIF antibody was conjugated to <i>p</i>-SCN-Bz-DFO and radiolabeled with positron-emitting zirconium-89 (<sup>89</sup>Zr). Target binding properties and stability of the radioimmunoconjugate were assessed by ELISA and size exclusion chromatography. The biodistribution of [<sup>89</sup>Zr]Zr-DFO-anti-LIF was evaluated by PET/CT imaging in an orthotopic glioblastoma mouse model with LIF-positive (GL261N) and LIF-negative (GL261N-CRISPR/LIF) tumors at 24, 48, and 72 hours post-administration. Tumor LIF levels were measured ex vivo by immunohistochemistry. Mass spectrometry determined 2.4 ± 0.3 chelators per antibody molecule. Competitive ELISA demonstrated unaltered affinity post-conjugation. Radiolabeling at a 1 MBq of <sup>89</sup>Zr per 5 μg of anti-LIF ratio achieved &gt;68% yield, &gt;95% purity, and 0.17 ± 0.03 MBq/μg specific activity. The radioimmunoconjugate remained &gt;90% intact after 72 h in both saline and mouse serum. PET imaging revealed specific accumulation in LIF-positive brain tumors in vivo (6 ± 1.26 % ID/mL at 72 h), which was 2-fold higher than that observed in the GL261N-CRISPR/LIF model. [<sup>89</sup>Zr]Zr-DFO-anti-LIF was successfully synthesized, exhibiting specificity and stability in vitro and in vivo, thus supporting its potential for glioblastoma monitoring, as well as guiding anti-LIF therapies.</p>","PeriodicalId":36426,"journal":{"name":"ACS Pharmacology and Translational Science","volume":"8 11","pages":"3886–3895"},"PeriodicalIF":3.7,"publicationDate":"2025-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145499662","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","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
ACS Pharmacology and Translational Science
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