首页 > 最新文献

Journal of Medicinal Chemistry最新文献

英文 中文
Efficacy and Toxicity Analysis of Selective BET Bromodomain Inhibitors in Models of Inflammatory Liver Disease
IF 7.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-04-14 DOI: 10.1021/acs.jmedchem.4c02555
Luke C. Doskey, Cole R. Scholtz, Nora R. Vail, Shalil Khanal, Amani L. Lee, Sai Giridhar Sarma Kandanur, Zachariah J. Hoell, Amelia M. Huehls, Mohamed R. Issa, Enis Kostallari, Sheng Cao, Joel M. Reid, Vijay H. Shah, Harmeet Malhi, William C. K. Pomerantz
BET bromodomain inhibitors demonstrate significant promise as anti-inflammatory agents. However, clinical data demonstrated that nonselective BET bromodomain inhibitors led to significant dose-limiting toxicity in clinical settings. Here, we use three orally bioavailable inhibitors, 13, that are either BRD4-D1 selective or pan-D1-biased + BRD4-D2, for assessing their cellular and in vivo efficacy and safety profile compared to known BET inhibitors in two inflammatory disease models. Our results show that pan-D1-biased + BRD4-D2 inhibitor, 3, is as efficacious as pan-BET inhibitor, I-BET151, in reducing inflammation in both models, whereas pan-D2 inhibitors are less effective. BRD4-D1 selective inhibitors are also efficacious; however, inhibitors with improved cellular engagement will be necessary to better assess their effects. Finally, BRD4-D1 selective inhibitors are better tolerated in a preclinical thrombocytopenia model than 3, while gastrointestinal toxicity may be a BRD4-driven effect. These results highlight the importance of assessing specific BET bromodomain functions due to their diverse roles in disease models.
{"title":"Efficacy and Toxicity Analysis of Selective BET Bromodomain Inhibitors in Models of Inflammatory Liver Disease","authors":"Luke C. Doskey, Cole R. Scholtz, Nora R. Vail, Shalil Khanal, Amani L. Lee, Sai Giridhar Sarma Kandanur, Zachariah J. Hoell, Amelia M. Huehls, Mohamed R. Issa, Enis Kostallari, Sheng Cao, Joel M. Reid, Vijay H. Shah, Harmeet Malhi, William C. K. Pomerantz","doi":"10.1021/acs.jmedchem.4c02555","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c02555","url":null,"abstract":"BET bromodomain inhibitors demonstrate significant promise as anti-inflammatory agents. However, clinical data demonstrated that nonselective BET bromodomain inhibitors led to significant dose-limiting toxicity in clinical settings. Here, we use three orally bioavailable inhibitors, <b>1</b>–<b>3</b>, that are either BRD4-D1 selective or pan-D1-biased + BRD4-D2, for assessing their cellular and in vivo efficacy and safety profile compared to known BET inhibitors in two inflammatory disease models. Our results show that pan-D1-biased + BRD4-D2 inhibitor, <b>3</b>, is as efficacious as pan-BET inhibitor, I-BET151, in reducing inflammation in both models, whereas pan-D2 inhibitors are less effective. BRD4-D1 selective inhibitors are also efficacious; however, inhibitors with improved cellular engagement will be necessary to better assess their effects. Finally, BRD4-D1 selective inhibitors are better tolerated in a preclinical thrombocytopenia model than <b>3</b>, while gastrointestinal toxicity may be a BRD4-driven effect. These results highlight the importance of assessing specific BET bromodomain functions due to their diverse roles in disease models.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"108 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-04-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143827250","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PCSK9 Targeted Autophagosome-Tethering Compounds: Design, Synthesis, and Antiatherosclerosis Evaluation
IF 7.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-04-14 DOI: 10.1021/acs.jmedchem.4c02915
Hongyu Wu, Ziwen Zhang, Yongxing Xue, Jiannan Guo, Zhirong Ouyang, Zhonglian Cao, Wei Guo, Qingwen Zhang, Mo Wang, Xianfeng Gu
Atherosclerosis is a multifaceted disease involving various cell types and complex mechanisms, and it is the main cause of cardiovascular disease. Proprotein convertase subtilisin/kexin type-9 (PCSK9) has been identified as an effective target for treating atherosclerosis; however, most current research focuses on biological drugs. Our work optimized the previously reported autophagosome-tethering compound OY3, and specifically, compound W6 induced PCSK9 degradation with a 5-fold increase in activity and a 6-fold increase in bioavailability. Compared to the currently marketed PCSK9 drug, siRNA, W6 demonstrated comparable antiatherosclerosis effects both in vivo and in vitro. W6 exhibited beneficial effects on hepatocytes, endothelial cells, macrophages, and vascular smooth muscle cells involved in the atherosclerosis process, making it a promising potential antiatherosclerosis drug. This work highlights the feasibility of ATTECs in degrading both intracellular and extracellular proteins, and our novel PCSK9-ATTEC W6 provides a valuable reference for the treatment of atherosclerotic diseases.
{"title":"PCSK9 Targeted Autophagosome-Tethering Compounds: Design, Synthesis, and Antiatherosclerosis Evaluation","authors":"Hongyu Wu, Ziwen Zhang, Yongxing Xue, Jiannan Guo, Zhirong Ouyang, Zhonglian Cao, Wei Guo, Qingwen Zhang, Mo Wang, Xianfeng Gu","doi":"10.1021/acs.jmedchem.4c02915","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c02915","url":null,"abstract":"Atherosclerosis is a multifaceted disease involving various cell types and complex mechanisms, and it is the main cause of cardiovascular disease. Proprotein convertase subtilisin/kexin type-9 (PCSK9) has been identified as an effective target for treating atherosclerosis; however, most current research focuses on biological drugs. Our work optimized the previously reported autophagosome-tethering compound <b>OY3</b>, and specifically, compound <b>W6</b> induced PCSK9 degradation with a 5-fold increase in activity and a 6-fold increase in bioavailability. Compared to the currently marketed PCSK9 drug, siRNA, <b>W6</b> demonstrated comparable antiatherosclerosis effects both <i>in vivo</i> and <i>in vitro</i>. <b>W6</b> exhibited beneficial effects on hepatocytes, endothelial cells, macrophages, and vascular smooth muscle cells involved in the atherosclerosis process, making it a promising potential antiatherosclerosis drug. This work highlights the feasibility of ATTECs in degrading both intracellular and extracellular proteins, and our novel PCSK9-ATTEC <b>W6</b> provides a valuable reference for the treatment of atherosclerotic diseases.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"16 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-04-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143827251","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Bifunctional Inhibition of Botulinum Neurotoxin A Protease: Unexpected Active Site Inhibition Enhances Covalent Targeting of an Allosteric Site
IF 6.8 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-04-13 DOI: 10.1021/acs.jmedchem.5c0041710.1021/acs.jmedchem.5c00417
Mingliang Lin, Nishant Karadkhelkar, Yanjie Li, Lisa M. Eubanks, William H. Tepp, Sabine Pellett and Kim D. Janda*, 

Botulinum neurotoxin (BoNT) is the most toxic protein known to man and a Tier 1 bioterrorism agent. Among its serotypes, BoNT/A possesses the greatest potency and persistence, as such strategies to counteract it are highly coveted. Bifunctional molecules incorporating both metal chelation and a covalent warhead have shown great potential for blunting BoNT/A LC’s toxicity/longevity. To further explore this idea, new warheads as well as zinc metal-chelating scaffolds were prepared and examined. The structure–activity relationship and kinetic analyses of these inhibitors challenged the standard protease assay leading to a new screening platform implemented and validated. Reconnaissance studies from this new screening platform delineated an unprecedented structural flexibility associated with BoNT/A’s enzyme pocket, which can be induced by a small molecule for enhanced allosteric target inhibition of the protease. The culmination of these findings offers previously unrealized opportunities for neutralizing the BoNT/A protease and thus future in vivo applications.

{"title":"Bifunctional Inhibition of Botulinum Neurotoxin A Protease: Unexpected Active Site Inhibition Enhances Covalent Targeting of an Allosteric Site","authors":"Mingliang Lin,&nbsp;Nishant Karadkhelkar,&nbsp;Yanjie Li,&nbsp;Lisa M. Eubanks,&nbsp;William H. Tepp,&nbsp;Sabine Pellett and Kim D. Janda*,&nbsp;","doi":"10.1021/acs.jmedchem.5c0041710.1021/acs.jmedchem.5c00417","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.5c00417https://doi.org/10.1021/acs.jmedchem.5c00417","url":null,"abstract":"<p >Botulinum neurotoxin (BoNT) is the most toxic protein known to man and a Tier 1 bioterrorism agent. Among its serotypes, BoNT/A possesses the greatest potency and persistence, as such strategies to counteract it are highly coveted. Bifunctional molecules incorporating both metal chelation and a covalent warhead have shown great potential for blunting BoNT/A LC’s toxicity/longevity. To further explore this idea, new warheads as well as zinc metal-chelating scaffolds were prepared and examined. The structure–activity relationship and kinetic analyses of these inhibitors challenged the standard protease assay leading to a new screening platform implemented and validated. Reconnaissance studies from this new screening platform delineated an unprecedented structural flexibility associated with BoNT/A’s enzyme pocket, which can be induced by a small molecule for enhanced allosteric target inhibition of the protease. The culmination of these findings offers previously unrealized opportunities for neutralizing the BoNT/A protease and thus future <i>in vivo</i> applications.</p>","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"68 8","pages":"8796–8816 8796–8816"},"PeriodicalIF":6.8,"publicationDate":"2025-04-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143863233","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Bifunctional Inhibition of Botulinum Neurotoxin A Protease: Unexpected Active Site Inhibition Enhances Covalent Targeting of an Allosteric Site
IF 7.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-04-13 DOI: 10.1021/acs.jmedchem.5c00417
Mingliang Lin, Nishant Karadkhelkar, Yanjie Li, Lisa M. Eubanks, William H. Tepp, Sabine Pellett, Kim D. Janda
Botulinum neurotoxin (BoNT) is the most toxic protein known to man and a Tier 1 bioterrorism agent. Among its serotypes, BoNT/A possesses the greatest potency and persistence, as such strategies to counteract it are highly coveted. Bifunctional molecules incorporating both metal chelation and a covalent warhead have shown great potential for blunting BoNT/A LC’s toxicity/longevity. To further explore this idea, new warheads as well as zinc metal-chelating scaffolds were prepared and examined. The structure–activity relationship and kinetic analyses of these inhibitors challenged the standard protease assay leading to a new screening platform implemented and validated. Reconnaissance studies from this new screening platform delineated an unprecedented structural flexibility associated with BoNT/A’s enzyme pocket, which can be induced by a small molecule for enhanced allosteric target inhibition of the protease. The culmination of these findings offers previously unrealized opportunities for neutralizing the BoNT/A protease and thus future in vivo applications.
{"title":"Bifunctional Inhibition of Botulinum Neurotoxin A Protease: Unexpected Active Site Inhibition Enhances Covalent Targeting of an Allosteric Site","authors":"Mingliang Lin, Nishant Karadkhelkar, Yanjie Li, Lisa M. Eubanks, William H. Tepp, Sabine Pellett, Kim D. Janda","doi":"10.1021/acs.jmedchem.5c00417","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.5c00417","url":null,"abstract":"Botulinum neurotoxin (BoNT) is the most toxic protein known to man and a Tier 1 bioterrorism agent. Among its serotypes, BoNT/A possesses the greatest potency and persistence, as such strategies to counteract it are highly coveted. Bifunctional molecules incorporating both metal chelation and a covalent warhead have shown great potential for blunting BoNT/A LC’s toxicity/longevity. To further explore this idea, new warheads as well as zinc metal-chelating scaffolds were prepared and examined. The structure–activity relationship and kinetic analyses of these inhibitors challenged the standard protease assay leading to a new screening platform implemented and validated. Reconnaissance studies from this new screening platform delineated an unprecedented structural flexibility associated with BoNT/A’s enzyme pocket, which can be induced by a small molecule for enhanced allosteric target inhibition of the protease. The culmination of these findings offers previously unrealized opportunities for neutralizing the BoNT/A protease and thus future <i>in vivo</i> applications.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"27 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-04-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143827253","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Novel Aryl Sulfonium Modification on Vancomycin to Tackle MRSA and VRE In Vitro and In Vivo through Dual Enhanced Cell-Wall and Membrane Inhibition
IF 6.8 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-04-12 DOI: 10.1021/acs.jmedchem.4c0302810.1021/acs.jmedchem.4c03028
Yuanyuan Xie, Xiaowen Wang, Taopeng Chang, Zhifu Chen, Youhong Luo, Jingwen Zhang, Hui Wang, Jinhua Dong*, Feifei Chen*, Jinyong Zhang* and Dongliang Guan*, 

Gram-positive superbugs resistant to methicillin and vancomycin pose a severe threat to global public health, urgently demanding novel therapeutic strategies. Herein, we rationally designed and synthesized vancomycin derivatives modified with diverse aryl sulfonium moieties to reactivate its antibacterial potency. By optimizing the sulfonium-based SAR, we got derivatives 2–3 orders of magnitude more active in vitro than vancomycin. Subsequently, preliminary toxicity evaluations for the optimal derivative, 7e, indicated a favorable therapeutic index, while pharmacokinetic assays revealed its good properties, suggesting great drug-like potential. Notably, 7e showed extremely potent in vivo protection efficacy by only a single-dose treatment in the challenging methicillin-resistant Staphylococcus aureus and VRE lethal sepsis mice models. Moreover, two independent and synergistic mechanisms of action were uncovered: membrane perturbation and enhanced cell wall biosynthesis inhibition. These findings revealed the unknown role of sulfonium strategy in vitro and in vivo and positioned 7e as a promising candidate for future development.

{"title":"Novel Aryl Sulfonium Modification on Vancomycin to Tackle MRSA and VRE In Vitro and In Vivo through Dual Enhanced Cell-Wall and Membrane Inhibition","authors":"Yuanyuan Xie,&nbsp;Xiaowen Wang,&nbsp;Taopeng Chang,&nbsp;Zhifu Chen,&nbsp;Youhong Luo,&nbsp;Jingwen Zhang,&nbsp;Hui Wang,&nbsp;Jinhua Dong*,&nbsp;Feifei Chen*,&nbsp;Jinyong Zhang* and Dongliang Guan*,&nbsp;","doi":"10.1021/acs.jmedchem.4c0302810.1021/acs.jmedchem.4c03028","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c03028https://doi.org/10.1021/acs.jmedchem.4c03028","url":null,"abstract":"<p >Gram-positive superbugs resistant to methicillin and vancomycin pose a severe threat to global public health, urgently demanding novel therapeutic strategies. Herein, we rationally designed and synthesized vancomycin derivatives modified with diverse aryl sulfonium moieties to reactivate its antibacterial potency. By optimizing the sulfonium-based SAR, we got derivatives 2–3 orders of magnitude more active in vitro than vancomycin. Subsequently, preliminary toxicity evaluations for the optimal derivative, <b>7e</b>, indicated a favorable therapeutic index, while pharmacokinetic assays revealed its good properties, suggesting great drug-like potential. Notably, <b>7e</b> showed extremely potent in vivo protection efficacy by only a single-dose treatment in the challenging methicillin-resistant <i>Staphylococcus aureus</i> and VRE lethal sepsis mice models. Moreover, two independent and synergistic mechanisms of action were uncovered: membrane perturbation and enhanced cell wall biosynthesis inhibition. These findings revealed the unknown role of sulfonium strategy in vitro and in vivo and positioned <b>7e</b> as a promising candidate for future development.</p>","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"68 8","pages":"8310–8329 8310–8329"},"PeriodicalIF":6.8,"publicationDate":"2025-04-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143863231","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Design and Synthesis of FR-β Targeting Chimeric Molecules for Reprogramming Tumor-Associated Macrophages Using 6-Substituted Pyrrolo[2,3-d]pyrimidines as Targeting Ligands
IF 6.8 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-04-12 DOI: 10.1021/acs.jmedchem.4c0299510.1021/acs.jmedchem.4c02995
Lixiao Guo, Binghao Guo, Yuqing Wang, Yuwei Zheng, Shuo Sun, Mengqi Wu, Yingao Li, Deguang Jiang, Ruijuan Xing, Zenghui Sun, Yan Fu, Xin Li, Yining Zhang*, Yi Liu* and Lei Wang*, 

Tumor-associated macrophages (TAMs) are highly plastic tumor-infiltrating immune cells. Their reprogramming has emerged as a pivotal strategy in antitumor immunotherapy. The TLR7/8 agonist, IMDQ, has significant potential for reprogramming macrophages but lacks target specificity. To address this challenge, we developed novel folate receptor beta (FR-β) targeting chimeric molecules using 6-substituted pyrrolo[2,3-d]pyrimidines as high-affinity ligands, which demonstrate superior FR-β targeting capability compared with classical folic acid. These molecules integrate the FR-β targeting moiety with IMDQ, marking the first application of this immunomodulator in targeted chimeric constructs. In vitro and in vivo studies demonstrated that our chimeric molecules selectively reprogrammed TAMs toward an immunostimulatory phenotype, reshaped the tumor microenvironment, and inhibited tumor progression without systemic toxicity. Given that TAM accumulation is prevalent across all solid tumors, our strategy of precisely targeting and reprogramming of TAMs is universally applicable to treating various types of cancers, a potent and effective strategy for antitumor immunotherapy.

{"title":"Design and Synthesis of FR-β Targeting Chimeric Molecules for Reprogramming Tumor-Associated Macrophages Using 6-Substituted Pyrrolo[2,3-d]pyrimidines as Targeting Ligands","authors":"Lixiao Guo,&nbsp;Binghao Guo,&nbsp;Yuqing Wang,&nbsp;Yuwei Zheng,&nbsp;Shuo Sun,&nbsp;Mengqi Wu,&nbsp;Yingao Li,&nbsp;Deguang Jiang,&nbsp;Ruijuan Xing,&nbsp;Zenghui Sun,&nbsp;Yan Fu,&nbsp;Xin Li,&nbsp;Yining Zhang*,&nbsp;Yi Liu* and Lei Wang*,&nbsp;","doi":"10.1021/acs.jmedchem.4c0299510.1021/acs.jmedchem.4c02995","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c02995https://doi.org/10.1021/acs.jmedchem.4c02995","url":null,"abstract":"<p >Tumor-associated macrophages (TAMs) are highly plastic tumor-infiltrating immune cells. Their reprogramming has emerged as a pivotal strategy in antitumor immunotherapy. The TLR7/8 agonist, <b>IMDQ</b>, has significant potential for reprogramming macrophages but lacks target specificity. To address this challenge, we developed novel folate receptor beta (FR-β) targeting chimeric molecules using 6-substituted pyrrolo[2,3-<i>d</i>]pyrimidines as high-affinity ligands, which demonstrate superior FR-β targeting capability compared with classical folic acid. These molecules integrate the FR-β targeting moiety with <b>IMDQ</b>, marking the first application of this immunomodulator in targeted chimeric constructs. In <i>vitro</i> and in <i>vivo</i> studies demonstrated that our chimeric molecules selectively reprogrammed TAMs toward an immunostimulatory phenotype, reshaped the tumor microenvironment, and inhibited tumor progression without systemic toxicity. Given that TAM accumulation is prevalent across all solid tumors, our strategy of precisely targeting and reprogramming of TAMs is universally applicable to treating various types of cancers, a potent and effective strategy for antitumor immunotherapy.</p>","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"68 8","pages":"8295–8309 8295–8309"},"PeriodicalIF":6.8,"publicationDate":"2025-04-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143863232","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Design and Synthesis of FR-β Targeting Chimeric Molecules for Reprogramming Tumor-Associated Macrophages Using 6-Substituted Pyrrolo[2,3-d]pyrimidines as Targeting Ligands
IF 7.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-04-12 DOI: 10.1021/acs.jmedchem.4c02995
Lixiao Guo, Binghao Guo, Yuqing Wang, Yuwei Zheng, Shuo Sun, Mengqi Wu, Yingao Li, Deguang Jiang, Ruijuan Xing, Zenghui Sun, Yan Fu, Xin Li, Yining Zhang, Yi Liu, Lei Wang
Tumor-associated macrophages (TAMs) are highly plastic tumor-infiltrating immune cells. Their reprogramming has emerged as a pivotal strategy in antitumor immunotherapy. The TLR7/8 agonist, IMDQ, has significant potential for reprogramming macrophages but lacks target specificity. To address this challenge, we developed novel folate receptor beta (FR-β) targeting chimeric molecules using 6-substituted pyrrolo[2,3-d]pyrimidines as high-affinity ligands, which demonstrate superior FR-β targeting capability compared with classical folic acid. These molecules integrate the FR-β targeting moiety with IMDQ, marking the first application of this immunomodulator in targeted chimeric constructs. In vitro and in vivo studies demonstrated that our chimeric molecules selectively reprogrammed TAMs toward an immunostimulatory phenotype, reshaped the tumor microenvironment, and inhibited tumor progression without systemic toxicity. Given that TAM accumulation is prevalent across all solid tumors, our strategy of precisely targeting and reprogramming of TAMs is universally applicable to treating various types of cancers, a potent and effective strategy for antitumor immunotherapy.
{"title":"Design and Synthesis of FR-β Targeting Chimeric Molecules for Reprogramming Tumor-Associated Macrophages Using 6-Substituted Pyrrolo[2,3-d]pyrimidines as Targeting Ligands","authors":"Lixiao Guo, Binghao Guo, Yuqing Wang, Yuwei Zheng, Shuo Sun, Mengqi Wu, Yingao Li, Deguang Jiang, Ruijuan Xing, Zenghui Sun, Yan Fu, Xin Li, Yining Zhang, Yi Liu, Lei Wang","doi":"10.1021/acs.jmedchem.4c02995","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c02995","url":null,"abstract":"Tumor-associated macrophages (TAMs) are highly plastic tumor-infiltrating immune cells. Their reprogramming has emerged as a pivotal strategy in antitumor immunotherapy. The TLR7/8 agonist, <b>IMDQ</b>, has significant potential for reprogramming macrophages but lacks target specificity. To address this challenge, we developed novel folate receptor beta (FR-β) targeting chimeric molecules using 6-substituted pyrrolo[2,3-<i>d</i>]pyrimidines as high-affinity ligands, which demonstrate superior FR-β targeting capability compared with classical folic acid. These molecules integrate the FR-β targeting moiety with <b>IMDQ</b>, marking the first application of this immunomodulator in targeted chimeric constructs. In <i>vitro</i> and in <i>vivo</i> studies demonstrated that our chimeric molecules selectively reprogrammed TAMs toward an immunostimulatory phenotype, reshaped the tumor microenvironment, and inhibited tumor progression without systemic toxicity. Given that TAM accumulation is prevalent across all solid tumors, our strategy of precisely targeting and reprogramming of TAMs is universally applicable to treating various types of cancers, a potent and effective strategy for antitumor immunotherapy.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"183 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-04-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143822950","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Novel Aryl Sulfonium Modification on Vancomycin to Tackle MRSA and VRE In Vitro and In Vivo through Dual Enhanced Cell-Wall and Membrane Inhibition
IF 7.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-04-12 DOI: 10.1021/acs.jmedchem.4c03028
Yuanyuan Xie, Xiaowen Wang, Taopeng Chang, Zhifu Chen, Youhong Luo, Jingwen Zhang, Hui Wang, Jinhua Dong, Feifei Chen, Jinyong Zhang, Dongliang Guan
Gram-positive superbugs resistant to methicillin and vancomycin pose a severe threat to global public health, urgently demanding novel therapeutic strategies. Herein, we rationally designed and synthesized vancomycin derivatives modified with diverse aryl sulfonium moieties to reactivate its antibacterial potency. By optimizing the sulfonium-based SAR, we got derivatives 2–3 orders of magnitude more active in vitro than vancomycin. Subsequently, preliminary toxicity evaluations for the optimal derivative, 7e, indicated a favorable therapeutic index, while pharmacokinetic assays revealed its good properties, suggesting great drug-like potential. Notably, 7e showed extremely potent in vivo protection efficacy by only a single-dose treatment in the challenging methicillin-resistant Staphylococcus aureus and VRE lethal sepsis mice models. Moreover, two independent and synergistic mechanisms of action were uncovered: membrane perturbation and enhanced cell wall biosynthesis inhibition. These findings revealed the unknown role of sulfonium strategy in vitro and in vivo and positioned 7e as a promising candidate for future development.
{"title":"Novel Aryl Sulfonium Modification on Vancomycin to Tackle MRSA and VRE In Vitro and In Vivo through Dual Enhanced Cell-Wall and Membrane Inhibition","authors":"Yuanyuan Xie, Xiaowen Wang, Taopeng Chang, Zhifu Chen, Youhong Luo, Jingwen Zhang, Hui Wang, Jinhua Dong, Feifei Chen, Jinyong Zhang, Dongliang Guan","doi":"10.1021/acs.jmedchem.4c03028","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c03028","url":null,"abstract":"Gram-positive superbugs resistant to methicillin and vancomycin pose a severe threat to global public health, urgently demanding novel therapeutic strategies. Herein, we rationally designed and synthesized vancomycin derivatives modified with diverse aryl sulfonium moieties to reactivate its antibacterial potency. By optimizing the sulfonium-based SAR, we got derivatives 2–3 orders of magnitude more active in vitro than vancomycin. Subsequently, preliminary toxicity evaluations for the optimal derivative, <b>7e</b>, indicated a favorable therapeutic index, while pharmacokinetic assays revealed its good properties, suggesting great drug-like potential. Notably, <b>7e</b> showed extremely potent in vivo protection efficacy by only a single-dose treatment in the challenging methicillin-resistant <i>Staphylococcus aureus</i> and VRE lethal sepsis mice models. Moreover, two independent and synergistic mechanisms of action were uncovered: membrane perturbation and enhanced cell wall biosynthesis inhibition. These findings revealed the unknown role of sulfonium strategy in vitro and in vivo and positioned <b>7e</b> as a promising candidate for future development.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"19 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-04-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143822932","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Correction to “Discovery of Novel 5-Cyano-3-phenylindole-Based LSD1/HDAC Dual Inhibitors for Colorectal Cancer Treatment” 对 "发现治疗结直肠癌的新型 5-Cyano-3-phenylindole-Based LSD1/HDAC 双抑制剂 "的更正
IF 7.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-04-11 DOI: 10.1021/acs.jmedchem.5c00900
Hui-Juan Zhu, Hui-Min Zhou, Xiao-Xiao Zhou, Shi-Jie Li, Meng-Jie Zheng, Zhen Xu, Wen-Jing Dai, Yi-Bo Ban, Meng-Yao Zhang, Yi-Zhe Zhang, Jia-Rui Lu, Yong-Tao Xu, Sai-Qi Wang, Xiao-Jing Shi, Ying-Chao Duan
In our study investigating the impact of target compound 20c alongside positive control drugs (ORY-1001 and SAHA) on the expression levels of HDAC and LSD1 substrate proteins─specifically, Ac-tubulin, Ac-H3, Ac-H4, H3K4me1, H3K4me2, and H3K4me3─we conducted a series of experiments on different tumor cell lines, including HCT-116 and HT-29 cells. Regrettably, during the assembly of Figure 7 in the original manuscript, we inadvertently used an incorrect image for H3K4me1 and H3 in Figure 7E. Upon a recent and thorough review of the original manuscript and raw data, we identified this error and have now replaced the incorrect image with the correct one in the revised Figure 7. All authors have agreed to the change. The corrected figure still demonstrates that compound 20c significantly upregulates the expression level of H3K4me1, and this correction do not alter the scientific conclusions of our study in any way. Figure 7. Compound 20c simultaneously inhibited LSD1 and HDAC activities in HCT-116 and HT-29 cells. (A-D) Western blot analysis of the effects of compound 20c on the expression levels of Ac-tubulin, Ac-H3, Ac-H4, and H3 in HCT-116 and HT-29 cells; (E–H) Western blot analysis of the effects of compound 20c on the expression levels of H3K4me1, H3K4me2, H3K4me3, LSD1, and H3 in HCT-116 and HT-29 cells; (I, J) CETSA of LSD1 with 20c at 5.0 μM in HCT-116 and HT-29 lysates for 6 h; (K) CETSA of LSD1 with 20c at different concentrations (1.25, 2.5, and 5.0 μM) in HCT-116 lysates for 6 h; (L) Western blot analysis of the effects of compound 24 on the levels of H3K4me1, H3K4me2, and Ac-H3. We sincerely regret any confusion this may have caused and appreciate the readers’ understanding as we take the necessary steps to ensure the accuracy and integrity of our research. This article has not yet been cited by other publications.
{"title":"Correction to “Discovery of Novel 5-Cyano-3-phenylindole-Based LSD1/HDAC Dual Inhibitors for Colorectal Cancer Treatment”","authors":"Hui-Juan Zhu, Hui-Min Zhou, Xiao-Xiao Zhou, Shi-Jie Li, Meng-Jie Zheng, Zhen Xu, Wen-Jing Dai, Yi-Bo Ban, Meng-Yao Zhang, Yi-Zhe Zhang, Jia-Rui Lu, Yong-Tao Xu, Sai-Qi Wang, Xiao-Jing Shi, Ying-Chao Duan","doi":"10.1021/acs.jmedchem.5c00900","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.5c00900","url":null,"abstract":"In our study investigating the impact of target compound <b>20c</b> alongside positive control drugs (ORY-1001 and SAHA) on the expression levels of HDAC and LSD1 substrate proteins─specifically, Ac-tubulin, Ac-H3, Ac-H4, H3K4me1, H3K4me2, and H3K4me3─we conducted a series of experiments on different tumor cell lines, including HCT-116 and HT-29 cells. Regrettably, during the assembly of Figure 7 in the original manuscript, we inadvertently used an incorrect image for H3K4me1 and H3 in Figure 7E. Upon a recent and thorough review of the original manuscript and raw data, we identified this error and have now replaced the incorrect image with the correct one in the revised Figure 7. All authors have agreed to the change. The corrected figure still demonstrates that compound <b>20c</b> significantly upregulates the expression level of H3K4me1, and this correction do not alter the scientific conclusions of our study in any way. Figure 7. Compound <b>20c</b> simultaneously inhibited LSD1 and HDAC activities in HCT-116 and HT-29 cells. (A-D) Western blot analysis of the effects of compound <b>20c</b> on the expression levels of Ac-tubulin, Ac-H3, Ac-H4, and H3 in HCT-116 and HT-29 cells; (E–H) Western blot analysis of the effects of compound <b>20c</b> on the expression levels of H3K4me1, H3K4me2, H3K4me3, LSD1, and H3 in HCT-116 and HT-29 cells; (I, J) CETSA of LSD1 with <b>20c</b> at 5.0 μM in HCT-116 and HT-29 lysates for 6 h; (K) CETSA of LSD1 with <b>20c</b> at different concentrations (1.25, 2.5, and 5.0 μM) in HCT-116 lysates for 6 h; (L) Western blot analysis of the effects of compound <b>24</b> on the levels of H3K4me1, H3K4me2, and Ac-H3. We sincerely regret any confusion this may have caused and appreciate the readers’ understanding as we take the necessary steps to ensure the accuracy and integrity of our research. This article has not yet been cited by other publications.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"108 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-04-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143820139","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Rational Design of Methylene Blue–Raloxifene Conjugates for Efficient Breast Tumor Elimination Triggered by ERα Degradation
IF 6.8 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-04-11 DOI: 10.1021/acs.jmedchem.5c0049010.1021/acs.jmedchem.5c00490
Yu Zhang, Qiying Yu, Ziwei Wang, Luolong Qing, Xiaoman Mo, Bing Liu, Yue’e Chai, Bingqiong Yu, Yongxi Dong, Weidong Pan*, Silong Zhang* and Huan He*, 

Small molecules capable of degrading estrogen receptor α (ERα) are of significant interest in breast cancer treatment. Herein, we rationally designed a series of ERα degraders (MR1MR3) by conjugating methylene blue, a bifunctional photosensitizer, with the raloxifene pharmacophore. The lead compound MR3 exhibited high affinity to ERα, and it can induce a complete depletion of ERα in MCF7 breast cancer cells after 660 nm irradiation (0.4 W/cm2) for 1 min. Owing to the ERα degradation merit, MR3 displayed a 45-fold boosted anticancer activity (IC50 = 0.55 μM) after irradiation. In the breast cancer xenograft mouse model, MR3 induced an obvious tumor regression (tumor growth inhibition = 118%), which was superior to that of the FDA-approved ERα degrader Faslodex. These important features make MR3 extremely intriguing for breast cancer treatment.

{"title":"Rational Design of Methylene Blue–Raloxifene Conjugates for Efficient Breast Tumor Elimination Triggered by ERα Degradation","authors":"Yu Zhang,&nbsp;Qiying Yu,&nbsp;Ziwei Wang,&nbsp;Luolong Qing,&nbsp;Xiaoman Mo,&nbsp;Bing Liu,&nbsp;Yue’e Chai,&nbsp;Bingqiong Yu,&nbsp;Yongxi Dong,&nbsp;Weidong Pan*,&nbsp;Silong Zhang* and Huan He*,&nbsp;","doi":"10.1021/acs.jmedchem.5c0049010.1021/acs.jmedchem.5c00490","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.5c00490https://doi.org/10.1021/acs.jmedchem.5c00490","url":null,"abstract":"<p >Small molecules capable of degrading estrogen receptor α (ERα) are of significant interest in breast cancer treatment. Herein, we rationally designed a series of ERα degraders (<b>MR1</b>–<b>MR3</b>) by conjugating methylene blue, a bifunctional photosensitizer, with the raloxifene pharmacophore. The lead compound <b>MR3</b> exhibited high affinity to ERα, and it can induce a complete depletion of ERα in MCF7 breast cancer cells after 660 nm irradiation (0.4 W/cm<sup>2</sup>) for 1 min. Owing to the ERα degradation merit, <b>MR3</b> displayed a 45-fold boosted anticancer activity (IC<sub>50</sub> = 0.55 μM) after irradiation. In the breast cancer xenograft mouse model, <b>MR3</b> induced an obvious tumor regression (tumor growth inhibition = 118%), which was superior to that of the FDA-approved ERα degrader <b>Faslodex</b>. These important features make <b>MR3</b> extremely intriguing for breast cancer treatment.</p>","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"68 8","pages":"8861–8872 8861–8872"},"PeriodicalIF":6.8,"publicationDate":"2025-04-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143863202","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Journal of Medicinal Chemistry
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:481959085
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