Pub Date : 2025-11-29DOI: 10.1016/j.ejmech.2025.118429
Tiago A. Duarte , Choa P. Ng , Jorge A.R. Salvador , Ludovico Pipito , Jennifer Greaves , Vânia M. Moreira
Neuroinflammation is a hallmark of many neurodegenerative diseases, including Alzheimer's, Parkinson's and Huntington's disease, multiple sclerosis, and infantile neuronal ceroid lipofuscinosis. Dynamic protein S-acylation, a reversible lipid post-translational modification, is an important regulator in these processes. S-acylation is catalysed by the zDHHC palmitoyl acyltransferases, and removal of the acyl groups is mediated by acyl-protein thioesterases. S-acylation controls the localisation, stability, and function of around 48 % of all proteins in the nervous system, including synaptic scaffolds, ion channels, immune receptors, and trafficking proteins. Moreover, dysregulated S-acylation contributes to synaptic loss, aberrant immune signalling, and neurodegeneration. This review examines proteins implicated in neuroinflammation with reported S-acylase or deacylase activity, outlines current knowledge on disease-related alterations in S-acylation, and assesses the therapeutic promise of available small-molecule modulators. Linking the activity of these enzymes with human disease highlights the potential of reversible S-acylation as a source of innovative targets for drug discovery in neuroinflammation.
{"title":"S-acylation and neuroinflammation: the therapeutic potential of zDHHC and deacylase modulation","authors":"Tiago A. Duarte , Choa P. Ng , Jorge A.R. Salvador , Ludovico Pipito , Jennifer Greaves , Vânia M. Moreira","doi":"10.1016/j.ejmech.2025.118429","DOIUrl":"10.1016/j.ejmech.2025.118429","url":null,"abstract":"<div><div>Neuroinflammation is a hallmark of many neurodegenerative diseases, including Alzheimer's, Parkinson's and Huntington's disease, multiple sclerosis, and infantile neuronal ceroid lipofuscinosis. Dynamic protein <em>S</em>-acylation, a reversible lipid post-translational modification, is an important regulator in these processes. <em>S</em>-acylation is catalysed by the zDHHC palmitoyl acyltransferases, and removal of the acyl groups is mediated by acyl-protein thioesterases. <em>S</em>-acylation controls the localisation, stability, and function of around 48 % of all proteins in the nervous system, including synaptic scaffolds, ion channels, immune receptors, and trafficking proteins. Moreover, dysregulated <em>S</em>-acylation contributes to synaptic loss, aberrant immune signalling, and neurodegeneration. This review examines proteins implicated in neuroinflammation with reported <em>S</em>-acylase or deacylase activity, outlines current knowledge on disease-related alterations in <em>S</em>-acylation, and assesses the therapeutic promise of available small-molecule modulators. Linking the activity of these enzymes with human disease highlights the potential of reversible <em>S</em>-acylation as a source of innovative targets for drug discovery in neuroinflammation.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"303 ","pages":"Article 118429"},"PeriodicalIF":5.9,"publicationDate":"2025-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145614076","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-29DOI: 10.1016/j.ejmech.2025.118408
Marzena Brinkmann , Tsung-Yun (Kent) Wong , Osha Roopnarine , Samantha L. Yuen , Kaja Berg , Razvan L. Cornea , Robyn T. Rebbeck , David D. Thomas , Courtney C. Aldrich
The sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2a) is a central regulator of cardiac Ca2+ handling and an emerging therapeutic target for heart failure. Here, we report a comprehensive structure–activity relationship (SAR) study around small-molecule activator compound 1, integrating Ca2+-ATPase and Ca2+-uptake assays, isoform selectivity profiling, and ADMET characterization across more than fifty analogues. Systematic modification of the left-hand aryl/heteroaryl region revealed a strong dependence of activity on aromaticity and lipophilicity, with CF3- and Br-substituted analogues providing substantial gains in potency. Optimization of the central amide linker established the importance of N-alkyl chain length, subtle hydrogen-bonding capacity, and a bent ligand geometry for productive SERCA2a engagement. Electronic tuning of the right-hand benzyl group further modulated efficacy, highlighting the essential contribution of an ortho-donor substituent. Functional evaluation across multiple Ca2+ concentrations identified several analogues with ATPase activation but inhibitory Ca2+-uptake effects, underscoring the need for dual-assay assessment to ensure bona fide activation. Among the series, compound 25 emerged as a balanced lead, displaying micromolar potency, robust concordant enhancement of ATPase and Ca2+-uptake activity, favorable solubility, and improved cytotoxicity relative to compound 1. Collectively, these findings define key structural determinants governing SERCA2a activation and provide a rational framework for developing next-generation, drug-like cardiac SERCA2a modulators.
{"title":"Development of activators for SERCA2a for heart failure treatments","authors":"Marzena Brinkmann , Tsung-Yun (Kent) Wong , Osha Roopnarine , Samantha L. Yuen , Kaja Berg , Razvan L. Cornea , Robyn T. Rebbeck , David D. Thomas , Courtney C. Aldrich","doi":"10.1016/j.ejmech.2025.118408","DOIUrl":"10.1016/j.ejmech.2025.118408","url":null,"abstract":"<div><div>The sarco/endoplasmic reticulum Ca<sup>2+</sup>-ATPase (SERCA2a) is a central regulator of cardiac Ca<sup>2+</sup> handling and an emerging therapeutic target for heart failure. Here, we report a comprehensive structure–activity relationship (SAR) study around small-molecule activator compound <strong>1</strong>, integrating Ca<sup>2+</sup>-ATPase and Ca<sup>2+</sup>-uptake assays, isoform selectivity profiling, and ADMET characterization across more than fifty analogues. Systematic modification of the left-hand aryl/heteroaryl region revealed a strong dependence of activity on aromaticity and lipophilicity, with CF<sub>3</sub>- and Br-substituted analogues providing substantial gains in potency. Optimization of the central amide linker established the importance of <em>N</em>-alkyl chain length, subtle hydrogen-bonding capacity, and a bent ligand geometry for productive SERCA2a engagement. Electronic tuning of the right-hand benzyl group further modulated efficacy, highlighting the essential contribution of an <em>ortho</em>-donor substituent. Functional evaluation across multiple Ca<sup>2+</sup> concentrations identified several analogues with ATPase activation but inhibitory Ca<sup>2+</sup>-uptake effects, underscoring the need for dual-assay assessment to ensure bona fide activation. Among the series, compound <strong>25</strong> emerged as a balanced lead, displaying micromolar potency, robust concordant enhancement of ATPase and Ca<sup>2+</sup>-uptake activity, favorable solubility, and improved cytotoxicity relative to compound <strong>1</strong>. Collectively, these findings define key structural determinants governing SERCA2a activation and provide a rational framework for developing next-generation, drug-like cardiac SERCA2a modulators.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"303 ","pages":"Article 118408"},"PeriodicalIF":5.9,"publicationDate":"2025-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145614056","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-29DOI: 10.1016/j.ejmech.2025.118394
Hao Xu , Dustin J.E. Huard , Elijah Dunn , Lucas A. Chalfoun , Felix Adulley , Raquel L. Lieberman , Brian S.J. Blagg
Grp94, the endoplasmic reticulum-resident paralog of Hsp90, is responsible for the folding and maturation of several client proteins including integrins and mutant myocilin. Inhibition of Grp94 with small molecules has been shown to reduce cell migration of breast cancer cells and promote degradation of mutant myocilin aggregates. Herein, we describe the development of 1,2,3-triazole based Grp94-selective inhibitors derived from a nitrogen scan on BnIm. Structure-activity relationship studies identified lead compound 47, which manifests 76 nM affinity for Grp94 with 121-fold selectivity over Hsp90α. In cellular studies, compound 47 induced the degradation of integrin α2 in MDA-MB-231 cells and reduced intracellular accumulation of mutant myocilin in human trabecular meshwork cells. These findings supported compound 47 as a potent and selective Grp94-selective inhibitor with therapeutic potential.
{"title":"Design, synthesis, and biological properties exhibited by 1,2,3-triazole based Grp94-selective inhibitors","authors":"Hao Xu , Dustin J.E. Huard , Elijah Dunn , Lucas A. Chalfoun , Felix Adulley , Raquel L. Lieberman , Brian S.J. Blagg","doi":"10.1016/j.ejmech.2025.118394","DOIUrl":"10.1016/j.ejmech.2025.118394","url":null,"abstract":"<div><div>Grp94, the endoplasmic reticulum-resident paralog of Hsp90, is responsible for the folding and maturation of several client proteins including integrins and mutant myocilin. Inhibition of Grp94 with small molecules has been shown to reduce cell migration of breast cancer cells and promote degradation of mutant myocilin aggregates. Herein, we describe the development of 1,2,3-triazole based Grp94-selective inhibitors derived from a nitrogen scan on <strong>BnIm</strong>. Structure-activity relationship studies identified lead compound <strong>47</strong>, which manifests 76 nM affinity for Grp94 with 121-fold selectivity over Hsp90α. In cellular studies, compound <strong>47</strong> induced the degradation of integrin α2 in MDA-MB-231 cells and reduced intracellular accumulation of mutant myocilin in human trabecular meshwork cells. These findings supported compound <strong>47</strong> as a potent and selective Grp94-selective inhibitor with therapeutic potential.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"303 ","pages":"Article 118394"},"PeriodicalIF":5.9,"publicationDate":"2025-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145614049","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-28DOI: 10.1016/j.ejmech.2025.118431
Runchu Li , Hongwei Lan , Jinyi Liu , Hui Gao, Yi Wei, Gaoteng Huang, Ying Wang, Yuxin Liu, Zihan Zhao, Li Wang, Wenjia Dan, Jiangkun Dai
The excessive and indiscriminate use of antibiotics has led to a broad increase in bacterial resistance, highlighting the critical need for the rapid development of new antibacterial agents. Herein, a collection of quaternary ammonium salts based on β-carboline skeleton was designed and prepared. Analysis of structure-activity relationships (SARs) revealed that the length of the hydrophobic chain is a critical determinant for antibacterial efficacy against methicillin-resistant Staphylococcus aureus (MRSA). Among them, compound 6d emerged as the most promising lead candidate, exhibiting strong activity against MRSA with MIC ranging from 0.25 to 0.5 μg/mL across 11 clinical isolates, while maintaining low hemolytic activity and minimal cytotoxicity toward mammalian cells. Furthermore, compound 6d displayed rapid bactericidal action at a high concentration (1 μg/mL), low propensity for inducing resistance, and good plasma stability. Notably, compound 6d demonstrated superior in vivo anti-MRSA activity and low toxicity. Further multi-target mechanistic studies indicated that compound 6d suppressed biofilm formation, compromised the cell wall, and disrupted the cytoplasmic membrane, which were accompanied by membrane depolarization, enhanced permeability, and loss of membrane integrity. Additionally, compound 6d facilitated the accumulation of reactive oxygen species (ROS), reduced metabolic activity as well as bound to DNA. Overall, these findings provide key insights for the development of quaternary ammonium salt antibacterial fusing active natural skeleton.
{"title":"Discovery of quaternary ammonium salts fusing β-carboline skeleton as new anti-MRSA agents","authors":"Runchu Li , Hongwei Lan , Jinyi Liu , Hui Gao, Yi Wei, Gaoteng Huang, Ying Wang, Yuxin Liu, Zihan Zhao, Li Wang, Wenjia Dan, Jiangkun Dai","doi":"10.1016/j.ejmech.2025.118431","DOIUrl":"10.1016/j.ejmech.2025.118431","url":null,"abstract":"<div><div>The excessive and indiscriminate use of antibiotics has led to a broad increase in bacterial resistance, highlighting the critical need for the rapid development of new antibacterial agents. Herein, a collection of quaternary ammonium salts based on <em>β</em>-carboline skeleton was designed and prepared. Analysis of structure-activity relationships (SARs) revealed that the length of the hydrophobic chain is a critical determinant for antibacterial efficacy against methicillin-resistant <em>Staphylococcus aureus</em> (MRSA). Among them, compound <strong>6d</strong> emerged as the most promising lead candidate, exhibiting strong activity against MRSA with MIC ranging from 0.25 to 0.5 μg/mL across 11 clinical isolates, while maintaining low hemolytic activity and minimal cytotoxicity toward mammalian cells. Furthermore, compound <strong>6d</strong> displayed rapid bactericidal action at a high concentration (1 μg/mL), low propensity for inducing resistance, and good plasma stability. Notably, compound <strong>6d</strong> demonstrated superior <em>in vivo</em> anti-MRSA activity and low toxicity. Further multi-target mechanistic studies indicated that compound <strong>6d</strong> suppressed biofilm formation, compromised the cell wall, and disrupted the cytoplasmic membrane, which were accompanied by membrane depolarization, enhanced permeability, and loss of membrane integrity. Additionally, compound <strong>6d</strong> facilitated the accumulation of reactive oxygen species (ROS), reduced metabolic activity as well as bound to DNA. Overall, these findings provide key insights for the development of quaternary ammonium salt antibacterial fusing active natural skeleton.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"303 ","pages":"Article 118431"},"PeriodicalIF":5.9,"publicationDate":"2025-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145611370","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-28DOI: 10.1016/j.ejmech.2025.118399
Chao Zhang , Yun-Sang Tang , Jian-Fei Gao , Zi-Xiao Liu , Shi-Shao Liang , Si-Miao You , Chris Ka-Pun Mok , Er-Fang Huang , Pang-Chui Shaw , Chun Hu
Novel imidazo[1,2-a]pyridine derivatives were designed as influenza A virus RNA-dependent RNA polymerase (RdRp) inhibitors via scaffold hybridization strategy. Forty-five synthesized compounds were screened by surface plasmon resonance (SPR) and bioactivity assays, which identified three compounds exhibiting potent antiviral activity against A/PR/8/34(H1N1) and strong binding to the target: 14 (IC50 = 3.00 μM; KD = 1.79 μM), 19 (IC50 = 0.95 μM; KD = 0.82 μM), and 41 (IC50 = 0.29 μM; KD = 4.11 μM). Moreover, compound 41 revealed significant broad-spectrum effects on multiple influenza virus strains. Structure-activity relationship (SAR) analysis identified key structural features on the imidazo[1,2-a]pyridine-3-carboxamide scaffold—including specific substitution patterns, linker types, and beneficial positions—that significantly enhanced inhibitory potency, providing a clear rationale for the development of potent influenza virus PA-PB1 inhibitors. Surface plasmon resonance analysis confirmed enhanced binding to the PAC domain, while molecular docking studies identified key interactions with LYS643 and GLN408 in PAC protein. Further molecular dynamics simulations and dynamic cross-correlation matrix analysis demonstrated stable binding modes and correlated motions within the PAC domain. In summary, compound 41 was identified as a promising sub-micromolar RdRp inhibitor targeting the PAC-PB1N interface. This study also showcases the applicability of SPR-affinity based screening approach in anti-influenza drug discovery.
{"title":"Discovery of imidazo[1,2-a]pyridine derivatives as potent anti-influenza agents: SPR affinity-based screening and structure-activity relationship study","authors":"Chao Zhang , Yun-Sang Tang , Jian-Fei Gao , Zi-Xiao Liu , Shi-Shao Liang , Si-Miao You , Chris Ka-Pun Mok , Er-Fang Huang , Pang-Chui Shaw , Chun Hu","doi":"10.1016/j.ejmech.2025.118399","DOIUrl":"10.1016/j.ejmech.2025.118399","url":null,"abstract":"<div><div>Novel imidazo[1,2-<em>a</em>]pyridine derivatives were designed as influenza A virus RNA-dependent RNA polymerase (RdRp) inhibitors via scaffold hybridization strategy. Forty-five synthesized compounds were screened by surface plasmon resonance (SPR) and bioactivity assays, which identified three compounds exhibiting potent antiviral activity against A/PR/8/34(H1N1) and strong binding to the target: <strong>14</strong> (IC<sub>50</sub> = 3.00 μM; <em>K</em><sub>D</sub> = 1.79 μM), <strong>19</strong> (IC<sub>50</sub> = 0.95 μM; <em>K</em><sub>D</sub> = 0.82 μM), and <strong>41</strong> (IC<sub>50</sub> = 0.29 μM; <em>K</em><sub>D</sub> = 4.11 μM). Moreover, compound <strong>41</strong> revealed significant broad-spectrum effects on multiple influenza virus strains. Structure-activity relationship (SAR) analysis identified key structural features on the imidazo[1,2-<em>a</em>]pyridine-3-carboxamide scaffold—including specific substitution patterns, linker types, and beneficial positions—that significantly enhanced inhibitory potency, providing a clear rationale for the development of potent influenza virus PA-PB1 inhibitors. Surface plasmon resonance analysis confirmed enhanced binding to the PA<sub>C</sub> domain, while molecular docking studies identified key interactions with LYS643 and GLN408 in PA<sub>C</sub> protein. Further molecular dynamics simulations and dynamic cross-correlation matrix analysis demonstrated stable binding modes and correlated motions within the PA<sub>C</sub> domain. In summary, compound <strong>41</strong> was identified as a promising sub-micromolar RdRp inhibitor targeting the PA<sub>C</sub>-PB1<sub>N</sub> interface. This study also showcases the applicability of SPR-affinity based screening approach in anti-influenza drug discovery.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"303 ","pages":"Article 118399"},"PeriodicalIF":5.9,"publicationDate":"2025-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145611368","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-28DOI: 10.1016/j.ejmech.2025.118402
Jiayue Pei , Caiyiren Wen , Qidong You , Lixiao Zhang , Yan Wang , Lei Wang
Heat shock protein 70 (HSP70), a crucial member of molecular chaperones, plays a pivotal role in maintaining cellular homeostasis and protecting cells from proteotoxic stress. Its strong pathophysiological associations with various malignancies, neurodegenerative disorders, cardiovascular diseases, and metabolic syndromes, establishing HSP70 as a promising therapeutic target. However, the clinical development of HSP70 inhibitors remains in its infancy, with only two small molecule inhibitors having entered to clinical trials. This limited clinical progress underscores the need for further optimization of current candidates and the development of novel HSP70-targeted inhibitors. This review summarizes the development of HSP70 inhibitors into three distinct chemical strategies: (1) pan-HSP70 inhibitors that target the conserved ATPase domain shared across isoforms; (2) isoform-selective inhibitors that address specific HSP70 family members; and (3) protein–protein interaction inhibitors that disrupt functional chaperone complexes. This review has systematically linked the structural analysis of HSP70 (including its structure and co-chaperone networks), disease-associated signaling pathways, and pathological roles to establish a classification framework based on its four major subtypes (HSP70i, HSC70, GRP78, and GRP75).
{"title":"Small molecules targeting HSP70 and therapeutic potentials","authors":"Jiayue Pei , Caiyiren Wen , Qidong You , Lixiao Zhang , Yan Wang , Lei Wang","doi":"10.1016/j.ejmech.2025.118402","DOIUrl":"10.1016/j.ejmech.2025.118402","url":null,"abstract":"<div><div>Heat shock protein 70 (HSP70), a crucial member of molecular chaperones, plays a pivotal role in maintaining cellular homeostasis and protecting cells from proteotoxic stress. Its strong pathophysiological associations with various malignancies, neurodegenerative disorders, cardiovascular diseases, and metabolic syndromes, establishing HSP70 as a promising therapeutic target. However, the clinical development of HSP70 inhibitors remains in its infancy, with only two small molecule inhibitors having entered to clinical trials. This limited clinical progress underscores the need for further optimization of current candidates and the development of novel HSP70-targeted inhibitors. This review summarizes the development of HSP70 inhibitors into three distinct chemical strategies: (1) pan-HSP70 inhibitors that target the conserved ATPase domain shared across isoforms; (2) isoform-selective inhibitors that address specific HSP70 family members; and (3) protein–protein interaction inhibitors that disrupt functional chaperone complexes. This review has systematically linked the structural analysis of HSP70 (including its structure and co-chaperone networks), disease-associated signaling pathways, and pathological roles to establish a classification framework based on its four major subtypes (HSP70i, HSC70, GRP78, and GRP75).</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"303 ","pages":"Article 118402"},"PeriodicalIF":5.9,"publicationDate":"2025-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145611424","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-28DOI: 10.1016/j.ejmech.2025.118413
Craig F. Steven, Manasa Punaha Ravindra, Martin Lee, Agata Makar, Christopher Stubbs, Pamela A. Lochhead, Poppy Winlow, Derek Barratt, Alex von Kriegsheim, Paul R.J. Davey, Valerie G. Brunton, Alison N. Hulme
Hydrophobic tagging (HyT) has emerged as a useful tool within the field of targeted protein degradation for the targeting and removal of a protein of interest (POI) from the cell. Development of new hydrophobic tags with increased efficiency not only allows increased POI degradation but also provides an opportunity to further study the mechanisms of HyT, which are not fully understood. Herein, we describe the development of new theranostic hydrophobic tags that can be used to induce protein degradation and simultaneously enable drug imaging using stimulated Raman scattering (SRS) microscopy. By coupling our dual-purpose theranostic hydrophobic tags to the PARP inhibiting, anti-cancer therapeutic olaparib, we explore the degradation efficiency, mode of action and potency that these theranostics confer in ovarian cancer cells. Lead compound 2c effectively degrades intracellular PARP-1 (DC50 = 0.618 μM, Dmax = 55%) and has more potent antiproliferative activity than the parent compound olaparib (IC50 = 62 nM vs 11.52 μM for olaparib), whilst maintaining on-target PARP inhibitory activity. Using mass spectrometry-based proteomic analysis, we demonstrate activation of the unfolded protein response, ER-stress and cell cycle arrest after incubation with 2c. Using SRS imaging, via the inherent Raman activity of the theranostic alkyne tag, we demonstrate involvement of the ER and autophagy after treatment with our drug conjugates. These results provide new insight into the mechanisms involved in HyT-induced protein degradation.
{"title":"New Mechanistic Insights into Targeted Protein Degradation Using Raman-Active Theranostic Hydrophobic Tags","authors":"Craig F. Steven, Manasa Punaha Ravindra, Martin Lee, Agata Makar, Christopher Stubbs, Pamela A. Lochhead, Poppy Winlow, Derek Barratt, Alex von Kriegsheim, Paul R.J. Davey, Valerie G. Brunton, Alison N. Hulme","doi":"10.1016/j.ejmech.2025.118413","DOIUrl":"https://doi.org/10.1016/j.ejmech.2025.118413","url":null,"abstract":"Hydrophobic tagging (HyT) has emerged as a useful tool within the field of targeted protein degradation for the targeting and removal of a protein of interest (POI) from the cell. Development of new hydrophobic tags with increased efficiency not only allows increased POI degradation but also provides an opportunity to further study the mechanisms of HyT, which are not fully understood. Herein, we describe the development of new theranostic hydrophobic tags that can be used to induce protein degradation and simultaneously enable drug imaging using stimulated Raman scattering (SRS) microscopy. By coupling our dual-purpose theranostic hydrophobic tags to the PARP inhibiting, anti-cancer therapeutic olaparib, we explore the degradation efficiency, mode of action and potency that these theranostics confer in ovarian cancer cells. Lead compound <strong>2c</strong> effectively degrades intracellular PARP-1 (DC<sub>50</sub> = 0.618 μM, D<sub>max</sub> = 55%) and has more potent antiproliferative activity than the parent compound olaparib (IC<sub>50</sub> = 62 nM vs 11.52 μM for olaparib), whilst maintaining on-target PARP inhibitory activity. Using mass spectrometry-based proteomic analysis, we demonstrate activation of the unfolded protein response, ER-stress and cell cycle arrest after incubation with <strong>2c</strong>. Using SRS imaging, via the inherent Raman activity of the theranostic alkyne tag, we demonstrate involvement of the ER and autophagy after treatment with our drug conjugates. These results provide new insight into the mechanisms involved in HyT-induced protein degradation.","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"8 1","pages":""},"PeriodicalIF":6.7,"publicationDate":"2025-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145611371","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-28DOI: 10.1016/j.ejmech.2025.118404
Alessia Costa , Gustavo Provensi , Costanza Titi , Manuela Leri , Monica Bucciantini , Marta Ferraroni , Adam B. Keeton , Austin M. Moore , Gary A. Piazza , Ashraf Hassan Abadi , Andrea Angeli , Claudiu T. Supuran
In this study, we report for the first time the design and evaluation of a series of compounds with potential therapeutic relevance for Alzheimer's disease (AD), able to inhibit both human Carbonic Anhydrase (hCA) isoforms most involved in this disease as well as Phosphodiesterase 5 (PDE5), using sildenafil as the structural scaffold. A total of 19 new dual-target molecules were synthesized and biologically assessed, leading to the identification of compound 8a as the most promising candidate, exhibiting potent inhibition toward both enzymatic targets. The binding interactions of three selected derivatives (6, 8a, and 10d) with hCA II were elucidated by X-ray crystallography experiments. Moreover, compound 8a demonstrated a favourable safety profile, as it did not markedly impair cell viability on differentiated SH-SY5Y at concentrations up to 100 μM and conferred protection against Aβ-induced cytotoxicity showing superior efficacy compared to the single-target reference agents acetazolamide (AAZ) and sildenafil in mitigating oxidative stress. In vivo, chronic administration of compound 8a prevented deficits in both recognition and working memory in Aβ1–42-infused mice, outperforming vehicle-treated controls. Collectively, these findings highlight the potential of dual CA/PDE5 inhibition as a novel therapeutic strategy for Alzheimer's disease.
{"title":"Exploring dual inhibitors Carbonic Anhydrases and Phosphodiesterase 5 as potential agents for treatment Alzheimer's disease","authors":"Alessia Costa , Gustavo Provensi , Costanza Titi , Manuela Leri , Monica Bucciantini , Marta Ferraroni , Adam B. Keeton , Austin M. Moore , Gary A. Piazza , Ashraf Hassan Abadi , Andrea Angeli , Claudiu T. Supuran","doi":"10.1016/j.ejmech.2025.118404","DOIUrl":"10.1016/j.ejmech.2025.118404","url":null,"abstract":"<div><div>In this study, we report for the first time the design and evaluation of a series of compounds with potential therapeutic relevance for Alzheimer's disease (AD), able to inhibit both human Carbonic Anhydrase (hCA) isoforms most involved in this disease as well as Phosphodiesterase 5 (PDE5), using sildenafil as the structural scaffold. A total of 19 new dual-target molecules were synthesized and biologically assessed, leading to the identification of compound <strong>8a</strong> as the most promising candidate, exhibiting potent inhibition toward both enzymatic targets. The binding interactions of three selected derivatives (<strong>6</strong>, <strong>8a</strong>, and <strong>10d</strong>) with hCA II were elucidated by X-ray crystallography experiments. Moreover, compound <strong>8a</strong> demonstrated a favourable safety profile, as it did not markedly impair cell viability on differentiated SH-SY5Y at concentrations up to 100 μM and conferred protection against Aβ-induced cytotoxicity showing superior efficacy compared to the single-target reference agents acetazolamide (AAZ) and sildenafil in mitigating oxidative stress. <em>In vivo</em>, chronic administration of compound <strong>8a</strong> prevented deficits in both recognition and working memory in Aβ<sub>1–42</sub>-infused mice, outperforming vehicle-treated controls. Collectively, these findings highlight the potential of dual CA/PDE5 inhibition as a novel therapeutic strategy for Alzheimer's disease.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"303 ","pages":"Article 118404"},"PeriodicalIF":5.9,"publicationDate":"2025-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145611369","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-27DOI: 10.1016/j.ejmech.2025.118398
Junying Song , Zhengyang He , Luyao Xie , Ailing Tian , Jinbu Xu , Jiao Tan , Feng Gao , Xiaohuan Li
Oridonin is an ent-kaurane diterpenoid with broader antitumor properties. Much efforts have been devoted to develop methods for rational chemical modification of oridonin to obtain synthetic derivatives with higher anticancer activity than the native structure. However, no study has examined the effect of stereochemistry on antiproliferative activity of oridonin and its derivatives thus far. In this study, a novel oridonin analogue with C14α-OH (6) was first synthesized via a Retro-Aldol-Aldol cascade reaction based on a configuration inversion strategy. Its structure was unambiguously confirmed by single-crystal X-ray diffraction analysis. The C14α-OH analogue (6), along with its native counterpart 2 (C14β-OH), was transformed into a series of forty-six C14–O-acylated products (6a-6w and 2a-2w). In vitro antiproliferative activities screening revealed that the α-configured derivative 6 (IC50 = 0.53 μM) exhibited 1.7-fold enhanced potency over its native β-form (2, IC50 = 0.88 μM) specifically in SU-DHL-6 cell lines, despite similar activity against solid tumors. The C14α-configured compounds 6k (IC50 = 0.12 μM) and 6l (IC50 = 0.31 μM) exhibited superior activity compared to their C14β-configured counterparts with the same substituents, 2k (IC50 = 0.71 μM) and 2l (IC50 = 0.47 μM), respectively. On the other hand, the aliphatic derivatives of C14α-O- series generally demonstrated reduced solubility, underscoring the importance of synergistic optimization of the substituent and configuration. Overall, the most potent compound 6k with a C14α-acrylate substitution (IC50 = 0.12 μM) demonstrated a 4.4-fold increase in potency compared to its parent compound 6 and was 16-fold more active than oridonin in the SU-DHL-6 cell lines. Mechanistic studies established that the most potent derivative 6k induces apoptosis and downregulates p-Akt in SU-DHL-6 cells without affecting cell cycle progression. Our study for the first time demonstrates that the stereochemistry at position C14 could affect the selectivity and antiproliferative activity of oridonin derivatives, providing novel natural product-derived lead compounds for further development of anti-cancer drugs.
{"title":"Effect of stereochemistry at position C14 on the antiproliferative activity and selectivity of simplified oridonin O-acylated derivatives","authors":"Junying Song , Zhengyang He , Luyao Xie , Ailing Tian , Jinbu Xu , Jiao Tan , Feng Gao , Xiaohuan Li","doi":"10.1016/j.ejmech.2025.118398","DOIUrl":"10.1016/j.ejmech.2025.118398","url":null,"abstract":"<div><div>Oridonin is an <em>ent</em>-kaurane diterpenoid with broader antitumor properties. Much efforts have been devoted to develop methods for rational chemical modification of oridonin to obtain synthetic derivatives with higher anticancer activity than the native structure. However, no study has examined the effect of stereochemistry on antiproliferative activity of oridonin and its derivatives thus far. In this study, a novel oridonin analogue with C14α-OH (<strong>6</strong>) was first synthesized via a Retro-Aldol-Aldol cascade reaction based on a configuration inversion strategy. Its structure was unambiguously confirmed by single-crystal X-ray diffraction analysis. The C14α-OH analogue (<strong>6</strong>), along with its native counterpart <strong>2</strong> (C14β-OH), was transformed into a series of forty-six C14–<em>O</em>-acylated products (<strong>6a-6w</strong> and <strong>2a-2w</strong>). In vitro antiproliferative activities screening revealed that the α-configured derivative <strong>6</strong> (IC<sub>50</sub> = 0.53 μM) exhibited 1.7-fold enhanced potency over its native β-form (<strong>2</strong>, IC<sub>50</sub> = 0.88 μM) specifically in SU-DHL-6 cell lines, despite similar activity against solid tumors. The C14α-configured compounds <strong>6k</strong> (IC<sub>50</sub> = 0.12 μM) and <strong>6l</strong> (IC<sub>50</sub> = 0.31 μM) exhibited superior activity compared to their C14β-configured counterparts with the same substituents, <strong>2k</strong> (IC<sub>50</sub> = 0.71 μM) and <strong>2l</strong> (IC<sub>50</sub> = 0.47 μM), respectively. On the other hand, the aliphatic derivatives of C14α-<em>O</em>- series generally demonstrated reduced solubility, underscoring the importance of synergistic optimization of the substituent and configuration. Overall, the most potent compound <strong>6k</strong> with a C14α-acrylate substitution (IC<sub>50</sub> = 0.12 μM) demonstrated a 4.4-fold increase in potency compared to its parent compound <strong>6</strong> and was 16-fold more active than oridonin in the SU-DHL-6 cell lines. Mechanistic studies established that the most potent derivative <strong>6k</strong> induces apoptosis and downregulates <em>p</em>-Akt in SU-DHL-6 cells without affecting cell cycle progression. Our study for the first time demonstrates that the stereochemistry at position C14 could affect the selectivity and antiproliferative activity of oridonin derivatives, providing novel natural product-derived lead compounds for further development of anti-cancer drugs.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"303 ","pages":"Article 118398"},"PeriodicalIF":5.9,"publicationDate":"2025-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145608856","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-27DOI: 10.1016/j.ejmech.2025.118395
Huanhuan Yin , Jiayin Long , Yukun Kong , Xiaoli Yin , Yiting Liu , Yujing Xu , Yujia Nie , Xiaoxue Liu , Yu Tang , Ximeng Shi , Tao An , Lei Ma
Microtubules have long been considered an ideal target for anticancer drugs because of the essential role they play in mitosis, forming the dynamic spindle apparatus. In this work, we reported the discovery of a novel series of homoisoflavonoids analogues bearing 3-benzylidene-6,7,8-trimethoxy-chroman-4-one moiety as tubulin polymerization inhibitors. The cytotoxic activity of the synthesized compounds was evaluated against two human cancer cell lines including A-375 and HL-60. Among them, compound B6 was identified as a potent tubulin polymerization inhibitor with effective anticancer potency, which demonstrated significant cytotoxic activity in vitro with IC50 values below 200 nM. Notably, it displayed remarkable inhibitory potency on A375 (IC50 = 90 nM) melanoma cancer cells, which surpassed those of CA-4 inhibitor. Mechanism explorations demonstrated that compound B6 inhibited tubulin polymerization by targeting the colchicine binding site, thereby disrupting the microtubule network in A375 cancer cells. Molecular docking studies of B6 into the colchicine-binding site of tubulin displayed possible mode of interaction between this compound and tubulin. Most anti-microtubule drugs inhibit cancer cell growth and proliferation by inducing cell cycle arrest during interphase, particularly at the G2/M phase. Additionally, the flow cytometry analysis of A375 and SK-MEL-28 cell lines treated with compound B6 showed that B6 effectively suppress colony formation, induce G2/M phase arrest, and promote apoptosis in A375 cancer cells, as well as regulate the expression levels of cell cycle and apoptosis-related proteins. Finally, the results of in vivo experiments indicated that the treatment with B6 could significantly inhibit tumor growth in a dose-dependent manner. Notably, the tumor weight growth inhibition (TGI) of B6 at 10 mg/kg was higher than that of CA-4 (15 mg/kg) and no significant body weight loss was observed for any of the treatments. All these results demonstrated that B6 showed in vivo anticancer activity and might be a promising hit compound.
{"title":"Synthesis and biological evaluation of polymethoxy homoisoflavanones as tubulin polymerization inhibitors","authors":"Huanhuan Yin , Jiayin Long , Yukun Kong , Xiaoli Yin , Yiting Liu , Yujing Xu , Yujia Nie , Xiaoxue Liu , Yu Tang , Ximeng Shi , Tao An , Lei Ma","doi":"10.1016/j.ejmech.2025.118395","DOIUrl":"10.1016/j.ejmech.2025.118395","url":null,"abstract":"<div><div>Microtubules have long been considered an ideal target for anticancer drugs because of the essential role they play in mitosis, forming the dynamic spindle apparatus. In this work, we reported the discovery of a novel series of homoisoflavonoids analogues bearing 3-benzylidene-6,7,8-trimethoxy-chroman-4-one moiety as tubulin polymerization inhibitors. The cytotoxic activity of the synthesized compounds was evaluated against two human cancer cell lines including A-375 and HL-60. Among them, compound <strong>B6</strong> was identified as a potent tubulin polymerization inhibitor with effective anticancer potency, which demonstrated significant cytotoxic activity <em>in vitro</em> with IC<sub>50</sub> values below 200 nM. Notably, it displayed remarkable inhibitory potency on A375 (IC<sub>50</sub> = 90 nM) melanoma cancer cells, which surpassed those of <strong>CA-4</strong> inhibitor. Mechanism explorations demonstrated that compound <strong>B6</strong> inhibited tubulin polymerization by targeting the colchicine binding site, thereby disrupting the microtubule network in A375 cancer cells. Molecular docking studies of <strong>B6</strong> into the colchicine-binding site of tubulin displayed possible mode of interaction between this compound and tubulin. Most anti-microtubule drugs inhibit cancer cell growth and proliferation by inducing cell cycle arrest during interphase, particularly at the G2/M phase. Additionally, the flow cytometry analysis of A375 and SK-MEL-28 cell lines treated with compound <strong>B6</strong> showed that <strong>B6</strong> effectively suppress colony formation, induce G2/M phase arrest, and promote apoptosis in A375 cancer cells, as well as regulate the expression levels of cell cycle and apoptosis-related proteins. Finally, the results of in vivo experiments indicated that the treatment with B6 could significantly inhibit tumor growth in a dose-dependent manner. Notably, the tumor weight growth inhibition (TGI) of B6 at 10 mg/kg was higher than that of CA-4 (15 mg/kg) and no significant body weight loss was observed for any of the treatments. All these results demonstrated that B6 showed in vivo anticancer activity and might be a promising hit compound.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"303 ","pages":"Article 118395"},"PeriodicalIF":5.9,"publicationDate":"2025-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145611415","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}