首页 > 最新文献

Medicinal Chemistry Research最新文献

英文 中文
How fast can you invent a medicine? 你能多快发明一种药?
IF 3.1 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-11-22 DOI: 10.1007/s00044-025-03501-6
Shireen R. Ashkar, Tim Cernak
{"title":"How fast can you invent a medicine?","authors":"Shireen R. Ashkar, Tim Cernak","doi":"10.1007/s00044-025-03501-6","DOIUrl":"10.1007/s00044-025-03501-6","url":null,"abstract":"","PeriodicalId":699,"journal":{"name":"Medicinal Chemistry Research","volume":"34 in","pages":"2365 - 2371"},"PeriodicalIF":3.1,"publicationDate":"2025-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145600903","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Special issue of Medicinal Chemistry Research in honor of Professor George A. Garcia 《药物化学研究》特刊纪念乔治·a·加西亚教授
IF 3.1 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-11-18 DOI: 10.1007/s00044-025-03502-5
Ashootosh Tripathi, Irina Artsimovitch
{"title":"Special issue of Medicinal Chemistry Research in honor of Professor George A. Garcia","authors":"Ashootosh Tripathi, Irina Artsimovitch","doi":"10.1007/s00044-025-03502-5","DOIUrl":"10.1007/s00044-025-03502-5","url":null,"abstract":"","PeriodicalId":699,"journal":{"name":"Medicinal Chemistry Research","volume":"34 in","pages":"2363 - 2364"},"PeriodicalIF":3.1,"publicationDate":"2025-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145600926","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Special issue of Medicinal Chemistry Research in honor of Professor Margaret O. James 《药物化学研究》特刊纪念玛格丽特·o·詹姆斯教授
IF 3.1 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-11-06 DOI: 10.1007/s00044-025-03497-z
Yousong Ding, Chengguo Xing, Hendrik Luesch
{"title":"Special issue of Medicinal Chemistry Research in honor of Professor Margaret O. James","authors":"Yousong Ding, Chengguo Xing, Hendrik Luesch","doi":"10.1007/s00044-025-03497-z","DOIUrl":"10.1007/s00044-025-03497-z","url":null,"abstract":"","PeriodicalId":699,"journal":{"name":"Medicinal Chemistry Research","volume":"34 in","pages":"2197 - 2199"},"PeriodicalIF":3.1,"publicationDate":"2025-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://link.springer.com/content/pdf/10.1007/s00044-025-03497-z.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145493359","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Rifamycin: discovery to future perspectives 利福霉素:从发现到未来的展望
IF 3.1 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-11-05 DOI: 10.1007/s00044-025-03491-5
Ashootosh Tripathi
{"title":"Rifamycin: discovery to future perspectives","authors":"Ashootosh Tripathi","doi":"10.1007/s00044-025-03491-5","DOIUrl":"10.1007/s00044-025-03491-5","url":null,"abstract":"","PeriodicalId":699,"journal":{"name":"Medicinal Chemistry Research","volume":"34 in","pages":"2372 - 2377"},"PeriodicalIF":3.1,"publicationDate":"2025-11-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145600925","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Anaeic and tumoic acids, new functionalized fatty acids from marine cyanobacteria, inhibit quorum sensing in Pseudomonas aeruginosa 贫血酸和肿瘤酸,新的功能化脂肪酸从海洋蓝藻,抑制群体感应铜绿假单胞菌
IF 3.1 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-11-05 DOI: 10.1007/s00044-025-03494-2
Emma K. Ellis, Sarath P. Gunasekera, Sofia Kokkaliari, Valerie J. Paul, Hendrik Luesch

Quorum sensing (QS) is the mechanism in which bacteria assess the threshold concentration of small signaling molecules to induce cell survival by overwhelming the host’s defenses. It is one of the fundamental mechanisms controlling bacterial mobility, motility, biofilm formation, and virulence. As many clinically resistant strains bypass traditional antimicrobial medications through this pathway, finding new therapeutics that target QS mechanisms is essential to treat these dangerous pathogens. Marine microbes are a promising source of potential therapeutics and have evolved molecules that can modulate disease-relevant signaling pathways, including QS. Compounds of various biosynthetic origin, including lipopeptides, alkaloids, cyclic peptides, and fatty acids have been shown to modulate QS pathways. In the present work, we have described the isolation, structure characterization, and biological evaluation of two new fatty acids, anaeic acid and tumoic acid. Anaeic acid (3) is a cyclopropanated fatty acid and shorter chain homologue of lyngbyoic acid (1). Tumoic acid (4) is the (6R)-methylated analogue of pitinoic acid A (2), characterized by a C-5 exo-double bond. The configurational assignment of 3 was conducted through analysis of the 1H and 2D NMR to establish the trans configuration of the cyclopropane ring using the NOE correlations, and optical rotation was used to determine the absolute configuration of 3 compared to 1. The absolute configuration of 4 was established through chemical degradation and derivatization of the liberated secondary alcohol with a chiral auxiliary, followed by Mosher’s ester analysis. Both compounds were compared directly with their close cyanobacterial analogues 1 and 2, distinguished by different chain length and differences in methylation, and their unbranched counterparts to assess the impact of the substitution pattern on functional responses with respect to QS. Compounds 1-4 exhibited moderate QS inhibitory activity against Pseudomonas aeruginosa based on the reduced virulence factor production and transcriptional response. In contrast, unbranched fatty acid counterparts octanoic (5), decanoic acid (6) and lauric acid (7) showed a propensity to activate QS signaling, although this effect was only significantly manifested on the transcriptional level.

群体感应(Quorum sensing, QS)是细菌评估小信号分子阈值浓度的机制,通过压倒宿主的防御来诱导细胞存活。它是控制细菌迁移、运动、生物膜形成和毒力的基本机制之一。由于许多临床耐药菌株通过这一途径绕过传统的抗微生物药物,因此寻找针对QS机制的新疗法对于治疗这些危险病原体至关重要。海洋微生物是潜在治疗药物的一个有希望的来源,并且已经进化出可以调节疾病相关信号通路的分子,包括QS。各种生物合成来源的化合物,包括脂肽、生物碱、环肽和脂肪酸,已被证明可以调节QS途径。在本工作中,我们描述了两种新的脂肪酸,厌氧酸和肿瘤酸的分离、结构表征和生物学评价。厌氧酸(3)是一种环丙化脂肪酸,是lynbyoic酸(1)的短链同源物。Tumoic acid(4)是pitinoic acid A(2)的(6R)甲基化类似物,以C-5外双键为特征。通过1H和2D NMR分析对3进行构型赋值,利用NOE相关性建立环丙烷环的反式构型,并利用旋光性确定3相对于1的绝对构型。通过手性助剂对释放的仲醇进行化学降解和衍生化,然后进行Mosher酯分析,确定了4的绝对构型。将这两种化合物与蓝藻类似物1和2进行直接比较,以评估取代模式对QS功能响应的影响。蓝藻类似物1和2具有不同的链长和甲基化差异。化合物1-4对铜绿假单胞菌表现出中等的QS抑制活性,这是基于减少毒力因子的产生和转录反应。相比之下,未支链脂肪酸对应物辛烷酸(5)、癸烷酸(6)和月桂酸(7)具有激活QS信号的倾向,尽管这种作用仅在转录水平上显著表现出来。
{"title":"Anaeic and tumoic acids, new functionalized fatty acids from marine cyanobacteria, inhibit quorum sensing in Pseudomonas aeruginosa","authors":"Emma K. Ellis,&nbsp;Sarath P. Gunasekera,&nbsp;Sofia Kokkaliari,&nbsp;Valerie J. Paul,&nbsp;Hendrik Luesch","doi":"10.1007/s00044-025-03494-2","DOIUrl":"10.1007/s00044-025-03494-2","url":null,"abstract":"<div><p>Quorum sensing (QS) is the mechanism in which bacteria assess the threshold concentration of small signaling molecules to induce cell survival by overwhelming the host’s defenses. It is one of the fundamental mechanisms controlling bacterial mobility, motility, biofilm formation, and virulence. As many clinically resistant strains bypass traditional antimicrobial medications through this pathway, finding new therapeutics that target QS mechanisms is essential to treat these dangerous pathogens. Marine microbes are a promising source of potential therapeutics and have evolved molecules that can modulate disease-relevant signaling pathways, including QS. Compounds of various biosynthetic origin, including lipopeptides, alkaloids, cyclic peptides, and fatty acids have been shown to modulate QS pathways. In the present work, we have described the isolation, structure characterization, and biological evaluation of two new fatty acids, anaeic acid and tumoic acid. Anaeic acid (<b>3</b>) is a cyclopropanated fatty acid and shorter chain homologue of lyngbyoic acid (<b>1</b>). Tumoic acid (<b>4</b>) is the (6<i>R</i>)-methylated analogue of pitinoic acid A (<b>2</b>), characterized by a C-5 exo-double bond. The configurational assignment of <b>3</b> was conducted through analysis of the <sup>1</sup>H and 2D NMR to establish the <i>trans</i> configuration of the cyclopropane ring using the NOE correlations, and optical rotation was used to determine the absolute configuration of <b>3</b> compared to <b>1</b>. The absolute configuration of <b>4</b> was established through chemical degradation and derivatization of the liberated secondary alcohol with a chiral auxiliary, followed by Mosher’s ester analysis. Both compounds were compared directly with their close cyanobacterial analogues <b>1</b> and <b>2</b>, distinguished by different chain length and differences in methylation, and their unbranched counterparts to assess the impact of the substitution pattern on functional responses with respect to QS. Compounds <b>1</b>-<b>4</b> exhibited moderate QS inhibitory activity against <i>Pseudomonas aeruginosa</i> based on the reduced virulence factor production and transcriptional response. In contrast, unbranched fatty acid counterparts octanoic (<b>5</b>), decanoic acid (<b>6</b>) and lauric acid (<b>7</b>) showed a propensity to activate QS signaling, although this effect was only significantly manifested on the transcriptional level.</p><div><figure><div><div><picture><source><img></source></picture></div></div></figure></div></div>","PeriodicalId":699,"journal":{"name":"Medicinal Chemistry Research","volume":"34 in","pages":"2350 - 2361"},"PeriodicalIF":3.1,"publicationDate":"2025-11-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://link.springer.com/content/pdf/10.1007/s00044-025-03494-2.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145493521","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
IND-enabling preclinical studies of [11C]COU, a trapped metabolite PET radiotracer for monoamine oxidase-B 单胺氧化酶- b捕获代谢物PET示踪剂[11C]COU的临床前研究
IF 3.1 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-11-05 DOI: 10.1007/s00044-025-03496-0
Madison S. Frazier, Tanpreet Kaur, Jenelle Stauff, Wade P. Winton, Bradford D. Henderson, Alexandra S. Dumond, Xia Shao, David M. Raffel, Kirk A. Frey, Michael R. Kilbourn, Allen F. Brooks, Peter J. H. Scott

[11C]COU is a trapped metabolite radiotracer for in vivo analysis of Monoamine Oxidase B activity using positron emission tomography (PET) imaging. [11C]COU has the potential to quantify astrocytosis in the early stages of Alzheimer’s disease, providing an earlier marker of disease than currently available for staging disease progression. Prior preclinical studies have demonstrated the efficacy of this radiotracer in preclinical imaging studies, warranting the translation for clinical evaluation. In this paper, we describe results of the requisite preclinical studies required to obtain approval for translation of [11C]COU into first-in-human studies. Development and validation of a production method that conforms to the quality requirements described in the US Pharmacopeia was accomplished, along with preclinical rodent studies to determine human radiation dose estimates and a single acute dose pharmacology and toxicology study to establish that an injected mass dose 100-fold higher than the proposed PET imaging dose was below the no-observed-adverse-effect level (NOAEL). The production method was validated in triplicate, yielding [11C]COU in sufficient radiochemical yield (9.3 ± 0.008%), radiochemical purity (99.2 ± 0.002%) and molar activity (165 ± 65 TBq/mmol) for routine clinical use, and providing a product that was sterile and met (or exceeded) all quality control requirements for human use. Dosimetric analysis determined that the effective human dose of [11C]COU is 0.005 mSv/MBq, also acceptable for clinical use. Lastly, no observable adverse effects were noted at 86 μg/kg in rodent toxicology studies (100x the proposed human dose). From these results we received approval to advance [11C]COU into clinical studies.

[11C]COU是一种捕获代谢物放射性示踪剂,用于使用正电子发射断层扫描(PET)成像分析单胺氧化酶B的体内活性。[11C]COU有可能量化阿尔茨海默病早期的星形细胞增多症,提供比目前可用的疾病进展分期更早的疾病标志物。先前的临床前研究已经证明了这种放射性示踪剂在临床前成像研究中的有效性,保证了临床评估的翻译。在本文中,我们描述了获得批准将[11C]COU转化为首次人体研究所需的临床前研究结果。符合美国药典质量要求的生产方法的开发和验证已经完成,同时进行了临床前啮齿动物研究,以确定人体辐射剂量估计,并进行了单次急性剂量药理学和毒理学研究,以确定比建议的PET成像剂量高100倍的注射质量剂量低于未观察到的不良反应水平(NOAEL)。该生产方法进行了三次验证,获得了[11C]COU的放射化学产率(9.3±0.008%)、放射化学纯度(99.2±0.002%)和摩尔活性(165±65 TBq/mmol),可用于常规临床使用,并提供了无菌且满足(或超过)所有质量控制要求的产品。剂量学分析确定[11C]COU的人体有效剂量为0.005mSv/MBq,也可用于临床。最后,在啮齿动物毒理学研究中,86 μg/kg(人体建议剂量的100倍)没有观察到不良反应。根据这些结果,我们获得批准将[11C]COU推进临床研究。
{"title":"IND-enabling preclinical studies of [11C]COU, a trapped metabolite PET radiotracer for monoamine oxidase-B","authors":"Madison S. Frazier,&nbsp;Tanpreet Kaur,&nbsp;Jenelle Stauff,&nbsp;Wade P. Winton,&nbsp;Bradford D. Henderson,&nbsp;Alexandra S. Dumond,&nbsp;Xia Shao,&nbsp;David M. Raffel,&nbsp;Kirk A. Frey,&nbsp;Michael R. Kilbourn,&nbsp;Allen F. Brooks,&nbsp;Peter J. H. Scott","doi":"10.1007/s00044-025-03496-0","DOIUrl":"10.1007/s00044-025-03496-0","url":null,"abstract":"<div><p>[<sup>11</sup>C]COU is a trapped metabolite radiotracer for in vivo analysis of Monoamine Oxidase B activity using positron emission tomography (PET) imaging. [<sup>11</sup>C]COU has the potential to quantify astrocytosis in the early stages of Alzheimer’s disease, providing an earlier marker of disease than currently available for staging disease progression. Prior preclinical studies have demonstrated the efficacy of this radiotracer in preclinical imaging studies, warranting the translation for clinical evaluation. In this paper, we describe results of the requisite preclinical studies required to obtain approval for translation of [<sup>11</sup>C]COU into first-in-human studies. Development and validation of a production method that conforms to the quality requirements described in the US Pharmacopeia was accomplished, along with preclinical rodent studies to determine human radiation dose estimates and a single acute dose pharmacology and toxicology study to establish that an injected mass dose 100-fold higher than the proposed PET imaging dose was below the no-observed-adverse-effect level (NOAEL). The production method was validated in triplicate, yielding [<sup>11</sup>C]COU in sufficient radiochemical yield (9.3 ± 0.008%), radiochemical purity (99.2 ± 0.002%) and molar activity (165 ± 65 TBq/mmol) for routine clinical use, and providing a product that was sterile and met (or exceeded) all quality control requirements for human use. Dosimetric analysis determined that the effective human dose of [<sup>11</sup>C]COU is 0.005 mSv/MBq, also acceptable for clinical use. Lastly, no observable adverse effects were noted at 86 μg/kg in rodent toxicology studies (100x the proposed human dose). From these results we received approval to advance [<sup>11</sup>C]COU into clinical studies.</p><div><figure><div><div><picture><source><img></source></picture></div></div></figure></div></div>","PeriodicalId":699,"journal":{"name":"Medicinal Chemistry Research","volume":"34 in","pages":"2537 - 2546"},"PeriodicalIF":3.1,"publicationDate":"2025-11-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12622565/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145547542","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Lenacapavir, a first-in-class, long-acting capsid inhibitor approved for both HIV-1 treatment and prevention Lenacapavir,一种一流的长效衣壳抑制剂,被批准用于HIV-1的治疗和预防
IF 3.1 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-10-31 DOI: 10.1007/s00044-025-03495-1
Zi Hui, Xiang-Yang Ye, Yuting Zhang, Longqin Hu, Tian Xie, Zhao-Qian Liu

Lenacapavir is a first-in-class, long-acting HIV-1 capsid inhibitor and represents a significant advancement in both the treatment and prevention of HIV-1 infection. It is highly effective against both wild-type and multidrug-resistant strains of the virus. It has a unique mechanism of action involving the disruption of HIV-1 capsid assembly and nuclear import, which ultimately prevents the virus from integrating its genetic material into the host cell’s DNA and hinders viral replication. This review covers lenacapavir’s discovery and development, with a focus on its target validation, lead optimization, chemical synthesis, pharmacokinetic properties, safety profile, and clinical applications.

Lenacapavir是一种一流的长效HIV-1衣壳抑制剂,在治疗和预防HIV-1感染方面取得了重大进展。它对病毒的野生型和耐多药毒株都非常有效。它具有独特的作用机制,包括破坏HIV-1衣壳组装和核输入,最终阻止病毒将其遗传物质整合到宿主细胞的DNA中,并阻碍病毒复制。本文综述了lenacapavir的发现和开发,重点介绍了它的靶点验证、先导物优化、化学合成、药代动力学特性、安全性和临床应用。
{"title":"Lenacapavir, a first-in-class, long-acting capsid inhibitor approved for both HIV-1 treatment and prevention","authors":"Zi Hui,&nbsp;Xiang-Yang Ye,&nbsp;Yuting Zhang,&nbsp;Longqin Hu,&nbsp;Tian Xie,&nbsp;Zhao-Qian Liu","doi":"10.1007/s00044-025-03495-1","DOIUrl":"10.1007/s00044-025-03495-1","url":null,"abstract":"<div><p>Lenacapavir is a first-in-class, long-acting HIV-1 capsid inhibitor and represents a significant advancement in both the treatment and prevention of HIV-1 infection. It is highly effective against both wild-type and multidrug-resistant strains of the virus. It has a unique mechanism of action involving the disruption of HIV-1 capsid assembly and nuclear import, which ultimately prevents the virus from integrating its genetic material into the host cell’s DNA and hinders viral replication. This review covers lenacapavir’s discovery and development, with a focus on its target validation, lead optimization, chemical synthesis, pharmacokinetic properties, safety profile, and clinical applications.</p><div><figure><div><div><picture><source><img></source></picture></div></div></figure></div></div>","PeriodicalId":699,"journal":{"name":"Medicinal Chemistry Research","volume":"34 in","pages":"2472 - 2485"},"PeriodicalIF":3.1,"publicationDate":"2025-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://link.springer.com/content/pdf/10.1007/s00044-025-03495-1.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145600933","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Synthesis of simplified didehydro-cortistatin A derivatives as anti-proliferative agents 简化二脱氢皮质抑素A衍生物的合成及其抗增殖作用
IF 3.1 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-10-30 DOI: 10.1007/s00044-025-03486-2
Jeremy S. Coleman, Larissa Costa de Almeida, Laura E. Hanold, Michael J. Ferracane, Hendrik Luesch, Jane V. Aldrich

The natural product cortistatin A and its derivative didehydro-cortistatin A exhibit potent biological activity in different disease states, indicating the potential utility of their derivatives as treatments for a variety of diseases. The synthesis of the unique ring system found in these compounds is challenging, and therefore we designed analogs with a conventional steroidal scaffold that retained the A-ring functionalities with the stereochemistries found in the natural product, building on a previous report of simplified didehydro-cortistatin A analogs. The steroidal derivatives were synthesized in 9 steps from prednisone with different isoquinoline isomers incorporated at C17 via a Stille coupling in the last step. The analogs exhibited antiproliferative activity in HCT 116 colon cancer cells with low micromolar potency (HCT 116 IC50 = 4.80–11.5 µM) and rapid onset. The methodology described here can be used to prepare additional simplified didehydro-cortistatin A analogs for future biological applications.

天然产物皮质抑素A及其衍生物二脱氢皮质抑素A在不同疾病状态下表现出强大的生物活性,表明其衍生物作为多种疾病治疗的潜在效用。在这些化合物中发现的独特环系统的合成是具有挑战性的,因此我们设计了具有传统甾体支架的类似物,保留了天然产物中发现的立体化学的a环功能,建立在先前简化的双脱氢皮质抑素a类似物的基础上。以强的松为原料,经9步合成甾体衍生物,最后一步通过Stille偶联在C17上加入不同的异喹啉异构体。这些类似物在HCT 116结肠癌细胞中表现出低微摩尔效价(HCT 116 IC50 = 4.80 ~ 11.5µM)和快速起效的抗增殖活性。本文描述的方法可用于制备其他简化的双脱氢皮质抑素A类似物,用于未来的生物学应用。
{"title":"Synthesis of simplified didehydro-cortistatin A derivatives as anti-proliferative agents","authors":"Jeremy S. Coleman,&nbsp;Larissa Costa de Almeida,&nbsp;Laura E. Hanold,&nbsp;Michael J. Ferracane,&nbsp;Hendrik Luesch,&nbsp;Jane V. Aldrich","doi":"10.1007/s00044-025-03486-2","DOIUrl":"10.1007/s00044-025-03486-2","url":null,"abstract":"<div><p>The natural product cortistatin A and its derivative didehydro-cortistatin A exhibit potent biological activity in different disease states, indicating the potential utility of their derivatives as treatments for a variety of diseases. The synthesis of the unique ring system found in these compounds is challenging, and therefore we designed analogs with a conventional steroidal scaffold that retained the A-ring functionalities with the stereochemistries found in the natural product, building on a previous report of simplified didehydro-cortistatin A analogs. The steroidal derivatives were synthesized in 9 steps from prednisone with different isoquinoline isomers incorporated at C17 via a Stille coupling in the last step. The analogs exhibited antiproliferative activity in HCT 116 colon cancer cells with low micromolar potency (HCT 116 IC<sub>50</sub> = 4.80–11.5 <i>µ</i>M) and rapid onset. The methodology described here can be used to prepare additional simplified didehydro-cortistatin A analogs for future biological applications.</p><div><figure><div><div><picture><source><img></source></picture></div></div></figure></div></div>","PeriodicalId":699,"journal":{"name":"Medicinal Chemistry Research","volume":"34 in","pages":"2292 - 2298"},"PeriodicalIF":3.1,"publicationDate":"2025-10-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145493356","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Triclosan concurrently inhibits UGT and CYP enzymes but shows high selectivity toward CYP2C19 三氯生同时抑制UGT和CYP酶,但对CYP2C19具有较高的选择性
IF 3.1 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-10-29 DOI: 10.1007/s00044-025-03489-z
Guo Zhong, Megha Chandrashekhar, Theresa Aliwarga

Understanding the clearance pathways of drug candidates and the fraction metabolized (fm) by drug-metabolizing enzymes is a major focus during drug discovery and development process. While selective cytochrome P450 (CYP) inhibitors are widely available, the lack of potent pan- uridine 5’-diphospho-glucuronosyltransferases (UGT) inhibitors with minimal cross-inhibition on CYP enzymes limits the ability to evaluate the contribution of UGT to drug clearance in vitro and in vivo. This study screened five potential inhibitors—triclosan, salicylamide, valproic acid, benzoic acid, and borneol—across twelve human UGT isoforms using human liver microsomes (HLM) and Supersome®. Triclosan emerged as a potent pan-UGT inhibitor, exhibiting IC50 values below 10 µM for all tested isoforms, ranging from 0.43–9.9 µM. However, triclosan also inhibited multiple CYP enzymes with IC50 values ranging from 0.12 to 22 µM. The concurrent inhibition of multiple CYP enzymes limits the application of triclosan as a selective tool compound for UGT reaction phenotyping. The kinetic analysis revealed noncompetitive inhibition of UGT1A3-mediated telmisartan glucuronidation by triclosan, where the other tested compounds failed to inhibit UGT1A3. Notably, triclosan demonstrated high selectivity and potency toward CYP2C19 (IC50 0.12 µM), suggesting its potential use in CYP2C19 reaction phenotyping in HLM. Additionally, triclosan selectively inhibited flavin-containing monooxygenase 3 (FMO3) but not FMO5.

了解候选药物的清除途径和药物代谢酶代谢的部分(fm)是药物发现和开发过程中的主要焦点。虽然选择性细胞色素P450 (CYP)抑制剂广泛可用,但缺乏对CYP酶具有最小交叉抑制的强效泛尿嘧啶5 ' -二磷酸葡萄糖醛酸转移酶(UGT)抑制剂,限制了评估UGT对体外和体内药物清除作用的能力。本研究利用人肝微粒体(HLM)和Supersome®筛选了5种潜在的抑制物——三氯生、水杨胺、丙戊酸、苯甲酸和冰片——用于12种人UGT同型异构体。三氯生是一种有效的泛ugt抑制剂,所有测试的同种异构体的IC50值均低于10µM,范围为0.43-9.9µM。然而,三氯生也抑制多种CYP酶,IC50值在0.12 ~ 22µM之间。同时抑制多种CYP酶限制了三氯生作为UGT反应表型的选择性工具化合物的应用。动力学分析显示,三氯生对UGT1A3介导的替米沙坦葡萄糖醛酸化具有非竞争性抑制作用,而其他被测化合物对UGT1A3没有抑制作用。值得注意的是,三氯生对CYP2C19表现出高选择性和效力(IC50为0.12µM),提示其在HLM中CYP2C19反应表型的潜在应用。此外,三氯生选择性地抑制含黄素单加氧酶3 (FMO3),而不抑制FMO5。
{"title":"Triclosan concurrently inhibits UGT and CYP enzymes but shows high selectivity toward CYP2C19","authors":"Guo Zhong,&nbsp;Megha Chandrashekhar,&nbsp;Theresa Aliwarga","doi":"10.1007/s00044-025-03489-z","DOIUrl":"10.1007/s00044-025-03489-z","url":null,"abstract":"<div><p>Understanding the clearance pathways of drug candidates and the fraction metabolized (f<sub>m</sub>) by drug-metabolizing enzymes is a major focus during drug discovery and development process. While selective cytochrome P450 (CYP) inhibitors are widely available, the lack of potent pan- uridine 5’-diphospho-glucuronosyltransferases (UGT) inhibitors with minimal cross-inhibition on CYP enzymes limits the ability to evaluate the contribution of UGT to drug clearance in vitro and in vivo. This study screened five potential inhibitors—triclosan, salicylamide, valproic acid, benzoic acid, and borneol—across twelve human UGT isoforms using human liver microsomes (HLM) and Supersome®. Triclosan emerged as a potent pan-UGT inhibitor, exhibiting IC<sub>50</sub> values below 10 µM for all tested isoforms, ranging from 0.43–9.9 µM. However, triclosan also inhibited multiple CYP enzymes with IC<sub>50</sub> values ranging from 0.12 to 22 µM. The concurrent inhibition of multiple CYP enzymes limits the application of triclosan as a selective tool compound for UGT reaction phenotyping. The kinetic analysis revealed noncompetitive inhibition of UGT1A3-mediated telmisartan glucuronidation by triclosan, where the other tested compounds failed to inhibit UGT1A3. Notably, triclosan demonstrated high selectivity and potency toward CYP2C19 (IC<sub>50</sub> 0.12 µM), suggesting its potential use in CYP2C19 reaction phenotyping in HLM. Additionally, triclosan selectively inhibited flavin-containing monooxygenase 3 (FMO3) but not FMO5.</p><div><figure><div><div><picture><source><img></source></picture></div></div></figure></div></div>","PeriodicalId":699,"journal":{"name":"Medicinal Chemistry Research","volume":"34 in","pages":"2338 - 2349"},"PeriodicalIF":3.1,"publicationDate":"2025-10-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://link.springer.com/content/pdf/10.1007/s00044-025-03489-z.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145493354","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Molecular hybridization strategy: design and synthesize carvacrol-based fibrate derivatives as novel lipid-lowering agents 分子杂交策略:设计和合成以卡瓦酚为基础的新型降脂剂贝特衍生物
IF 3.1 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-10-27 DOI: 10.1007/s00044-025-03490-6
Yunbi Zhang, Wenjing Li, Kexin Xu, Boling Zhou, Huanxian Shi, Ling Ding, Huizi Shangguan, Yongheng Shi, Xinya Xu, Jiping Liu, Yundong Xie

This study was designed to synthesize a series of carvacrol-based fibrate derivatives based on a molecular hybridization strategy. In acute hyperlipidemic mice, the target compound T7 exhibited a noticeable effect on lowering lipid and demonstrated a dose-dependent characteristic. In the high-fat diet (HFD) mouse model, T7 notably decreased triglyceride (TG), total cholesterol (TC), and low-density lipoprotein cholesterol (LDL-C) levels, and elevated high-density lipoprotein cholesterol (HDL-C) levels in both serum and liver tissues. Additionally, T7 appreciably increased the aspartate aminotransferase (AST) and alanine aminotransferase (ALT) found in both serum and liver tissues. Liver histopathological examination indicated that it could inhibit hepatic lipid accumulation and alleviate liver injury. After administration, T7 exhibited anti-oxidative stress and anti-inflammatory effects. It could appreciably increase the activity of superoxide dismutase (SOD), decrease the activity of lipid peroxidation product malondialdehyde (MDA), and appreciably reduce the pro-inflammatory factors interleukin-6 (IL-6) and tumour necrosis factor-α (TNF-α) levels. Molecular docking experiments demonstrated that T7 exhibited a strong binding affinity with the peroxisome proliferator-activated receptor-α (PPAR-α) receptor. T7 enhanced the PPAR-α expression in liver tissues, indicating that T7 may regulate lipid metabolism by activating the PPAR-α receptor. The hepatoprotective effect of T7 may be closely linked to its capacity to reduce oxidative stress and inflammatory responses. In conclusion, T7 may be a potential novel lipid-lowering candidate compound with the potential to improve liver injury.

本研究旨在基于分子杂交策略合成一系列以香芹酚为基础的贝特衍生物。在急性高脂血症小鼠中,靶化合物T7表现出明显的降脂作用,并表现出剂量依赖性。在高脂饮食(HFD)小鼠模型中,T7显著降低血清和肝脏组织中甘油三酯(TG)、总胆固醇(TC)和低密度脂蛋白胆固醇(LDL-C)水平,升高高密度脂蛋白胆固醇(HDL-C)水平。此外,T7显著提高了血清和肝组织中谷草转氨酶(AST)和丙氨酸转氨酶(ALT)的含量。肝组织病理学检查显示其能抑制肝脏脂质积累,减轻肝损伤。给药后,T7表现出抗氧化应激和抗炎作用。能显著提高超氧化物歧化酶(SOD)活性,降低脂质过氧化产物丙二醛(MDA)活性,显著降低促炎因子白细胞介素-6 (IL-6)和肿瘤坏死因子-α (TNF-α)水平。分子对接实验表明,T7与过氧化物酶体增殖激活受体-α (PPAR-α)受体具有较强的结合亲和力。T7增强肝组织PPAR-α表达,提示T7可能通过激活PPAR-α受体调节脂质代谢。T7的肝保护作用可能与其减少氧化应激和炎症反应的能力密切相关。总之,T7可能是一种潜在的新型降脂候选化合物,具有改善肝损伤的潜力。
{"title":"Molecular hybridization strategy: design and synthesize carvacrol-based fibrate derivatives as novel lipid-lowering agents","authors":"Yunbi Zhang,&nbsp;Wenjing Li,&nbsp;Kexin Xu,&nbsp;Boling Zhou,&nbsp;Huanxian Shi,&nbsp;Ling Ding,&nbsp;Huizi Shangguan,&nbsp;Yongheng Shi,&nbsp;Xinya Xu,&nbsp;Jiping Liu,&nbsp;Yundong Xie","doi":"10.1007/s00044-025-03490-6","DOIUrl":"10.1007/s00044-025-03490-6","url":null,"abstract":"<div><p>This study was designed to synthesize a series of carvacrol-based fibrate derivatives based on a molecular hybridization strategy. In acute hyperlipidemic mice, the target compound T7 exhibited a noticeable effect on lowering lipid and demonstrated a dose-dependent characteristic. In the high-fat diet (HFD) mouse model, T7 notably decreased triglyceride (TG), total cholesterol (TC), and low-density lipoprotein cholesterol (LDL-C) levels, and elevated high-density lipoprotein cholesterol (HDL-C) levels in both serum and liver tissues. Additionally, T7 appreciably increased the aspartate aminotransferase (AST) and alanine aminotransferase (ALT) found in both serum and liver tissues. Liver histopathological examination indicated that it could inhibit hepatic lipid accumulation and alleviate liver injury. After administration, T7 exhibited anti-oxidative stress and anti-inflammatory effects. It could appreciably increase the activity of superoxide dismutase (SOD), decrease the activity of lipid peroxidation product malondialdehyde (MDA), and appreciably reduce the pro-inflammatory factors interleukin-6 (IL-6) and tumour necrosis factor-α (TNF-α) levels. Molecular docking experiments demonstrated that T7 exhibited a strong binding affinity with the peroxisome proliferator-activated receptor-α (PPAR-α) receptor. T7 enhanced the PPAR-α expression in liver tissues, indicating that T7 may regulate lipid metabolism by activating the PPAR-α receptor. The hepatoprotective effect of T7 may be closely linked to its capacity to reduce oxidative stress and inflammatory responses. In conclusion, T7 may be a potential novel lipid-lowering candidate compound with the potential to improve liver injury.</p><div><figure><div><div><picture><source><img></source></picture></div></div></figure></div></div>","PeriodicalId":699,"journal":{"name":"Medicinal Chemistry Research","volume":"34 in","pages":"2628 - 2646"},"PeriodicalIF":3.1,"publicationDate":"2025-10-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145600927","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Medicinal Chemistry Research
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