Pub Date : 2025-07-11DOI: 10.1021/acs.jmedchem.5c00919
Leon Katzengruber,Pascal Sander,Stefan Zwirner,Alexander Rasch,Eric Eberlein,Roland Selig,Wolfgang Albrecht,Lars Zender,Stefan A Laufer
Due to limited treatment options, liver failure remains a major challenge in modern medicine. With the validation of mitogen-activated protein kinase kinase 4 (MKK4, also known as MEK4 or MAP2K4) as a regulator of hepatocyte regeneration, a promising target for curative treatment of degenerative liver diseases was recently identified via in vivo RNAi experiments. The field of small molecules targeting MKK4 is of growing interest. Several MKK4 inhibitors with differing scaffolds are known, but few have reasonable selectivity profiles and drug-like properties. To further explore the space of drug-like MKK4 scaffolds, we performed a broad screening campaign and identified BI-D1870 as a promising candidate. The dihydropteridinone BI-D1870 is an unselective ribosomal S6 kinase inhibitor with broad off-target activity. In the study presented herein, we report a successful off-to-on target strategy that led to the development of highly selective 1,4-dihydropyrido[3,4-b]pyrazin-3(2H)-one inhibitors of MKK4.
{"title":"Discovery of the First Highly Selective 1,4-dihydropyrido[3,4-b]pyrazin-3(2H)-one MKK4 Inhibitor.","authors":"Leon Katzengruber,Pascal Sander,Stefan Zwirner,Alexander Rasch,Eric Eberlein,Roland Selig,Wolfgang Albrecht,Lars Zender,Stefan A Laufer","doi":"10.1021/acs.jmedchem.5c00919","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.5c00919","url":null,"abstract":"Due to limited treatment options, liver failure remains a major challenge in modern medicine. With the validation of mitogen-activated protein kinase kinase 4 (MKK4, also known as MEK4 or MAP2K4) as a regulator of hepatocyte regeneration, a promising target for curative treatment of degenerative liver diseases was recently identified via in vivo RNAi experiments. The field of small molecules targeting MKK4 is of growing interest. Several MKK4 inhibitors with differing scaffolds are known, but few have reasonable selectivity profiles and drug-like properties. To further explore the space of drug-like MKK4 scaffolds, we performed a broad screening campaign and identified BI-D1870 as a promising candidate. The dihydropteridinone BI-D1870 is an unselective ribosomal S6 kinase inhibitor with broad off-target activity. In the study presented herein, we report a successful off-to-on target strategy that led to the development of highly selective 1,4-dihydropyrido[3,4-b]pyrazin-3(2H)-one inhibitors of MKK4.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"4 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-07-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144603908","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}
Designing membrane-permeable drugs requires a precise understanding of noncovalent interactions governing cellular uptake. We propose a molecular thermodynamic–dynamic (MTD) framework that quantifies interaction thresholds dictating permeation efficiency, using polychlorinated biphenyls (PCBs) as structurally tunable probes. Our results reveal that optimal permeability occurs within a defined differential binding energy (ΔG = −3.6 to −6.8 kcal/mol for H-/X-bonding), facilitating membrane translocation through a binding-flip mechanism. Beyond this range, excessive binding affinity (ΔG < −7.5 kcal/mol) leads to kinetic trapping at the membrane surface. Notably, the membrane permeation coefficients exhibit a strong linear correlation with differential binding energy (R2 = 0.93), as revealed by five distinct transition states, including a rate-limiting vertical rotation step (ΔG = 2.4 kcal/mol). These findings yield two critical design principles: (i) intermediate differential binding (−4.0 to −5.0 kcal/mol) maximizes permeability, aligning with optimal ranges in FDA-approved membrane-permeable drugs, and (ii) targeted X-bonding modulation precisely controls membrane interaction specificity.
{"title":"Noncovalent Interaction Thresholds Control Translocation and Cytotoxicity: A Combined Computational–Experimental Study","authors":"Xianyu Song, Xianli Duan, Wenjun Xiang, Shuangliang Zhao","doi":"10.1021/acs.jmedchem.5c01196","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.5c01196","url":null,"abstract":"Designing membrane-permeable drugs requires a precise understanding of noncovalent interactions governing cellular uptake. We propose a molecular thermodynamic–dynamic (MTD) framework that quantifies interaction thresholds dictating permeation efficiency, using polychlorinated biphenyls (PCBs) as structurally tunable probes. Our results reveal that optimal permeability occurs within a defined differential binding energy (Δ<i>G</i> = −3.6 to −6.8 kcal/mol for H-/X-bonding), facilitating membrane translocation through a binding-flip mechanism. Beyond this range, excessive binding affinity (Δ<i>G</i> < −7.5 kcal/mol) leads to kinetic trapping at the membrane surface. Notably, the membrane permeation coefficients exhibit a strong linear correlation with differential binding energy (<i>R</i><sup>2</sup> = 0.93), as revealed by five distinct transition states, including a rate-limiting vertical rotation step (Δ<i>G</i> = 2.4 kcal/mol). These findings yield two critical design principles: (i) intermediate differential binding (−4.0 to −5.0 kcal/mol) maximizes permeability, aligning with optimal ranges in FDA-approved membrane-permeable drugs, and (ii) targeted X-bonding modulation precisely controls membrane interaction specificity.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"12 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-07-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144603907","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}
Polarity changes are significant indicators during inflammation. Acute lung injury (ALI) and asthma, both inflammatory lung diseases, necessitate the detection of these polarity changes for early diagnosis and treatment monitoring. We designed and synthesized a lipid droplet-targeting, polarity-dependent near-infrared fluorescent probe, MY-1, on the basis of an intramolecular charge-transfer mechanism to noninvasively detect these inflammatory conditions. The probe exhibits a consistent decrease in fluorescence intensity with an increase in solvent polarity, demonstrating excellent photostability and biocompatibility. Utilizing MY-1, we can differentiate between normal cells and cancer cells and successfully detect polarity changes targeting lipid droplets within the lungs of mice suffering from ALI and asthma. Furthermore, we can monitor the therapeutic effects of dexamethasone on asthmatic mice, confirming the clinical application potential of MY-1. In summary, we believe that MY-1 offers a novel and effective approach to the diagnosis of acute ALI and asthma.
{"title":"Lipid Droplet-Targeted NIR Polarity Probe for the Imaging of Acute Asthma In Vivo.","authors":"Mingyue Li,Yanmei Zhu,Linlin Hu,Jun Li,Jiyoung Yoo,Huamei Zhang,Zhaosheng Qian,Xucong Zhou,Jong Seung Kim,Jin Zhou","doi":"10.1021/acs.jmedchem.5c01403","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.5c01403","url":null,"abstract":"Polarity changes are significant indicators during inflammation. Acute lung injury (ALI) and asthma, both inflammatory lung diseases, necessitate the detection of these polarity changes for early diagnosis and treatment monitoring. We designed and synthesized a lipid droplet-targeting, polarity-dependent near-infrared fluorescent probe, MY-1, on the basis of an intramolecular charge-transfer mechanism to noninvasively detect these inflammatory conditions. The probe exhibits a consistent decrease in fluorescence intensity with an increase in solvent polarity, demonstrating excellent photostability and biocompatibility. Utilizing MY-1, we can differentiate between normal cells and cancer cells and successfully detect polarity changes targeting lipid droplets within the lungs of mice suffering from ALI and asthma. Furthermore, we can monitor the therapeutic effects of dexamethasone on asthmatic mice, confirming the clinical application potential of MY-1. In summary, we believe that MY-1 offers a novel and effective approach to the diagnosis of acute ALI and asthma.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"303 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-07-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144603910","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}
Yong Ju, Huizhu Song, Yuelan He, Yingtung Lo, Zhipeng Fan and Jianzhong Lu*,
{"title":"","authors":"Yong Ju, Huizhu Song, Yuelan He, Yingtung Lo, Zhipeng Fan and Jianzhong Lu*, ","doi":"","DOIUrl":"","url":null,"abstract":"","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"68 13","pages":"XXX-XXX 12260–12268"},"PeriodicalIF":6.8,"publicationDate":"2025-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/pdf/10.1021/acs.jmedchem.5c01022","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144587369","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-07-10Epub Date: 2025-06-13DOI: 10.1021/acs.jmedchem.5c00809
Ranjan Kumar Acharyya, Liyue Huang, Angelo Aguilar, Biao Hu, Longchuan Bai, Hoda Metwally, Donna McEachern, Wei Jiang, Yu Wang, Qiuxia Li, Bo Wen, Duxin Sun, Shaomeng Wang
MDM2 is a key negative regulator of the tumor suppressor p53 and an attractive target for cancer therapy. We report the discovery of MD-4251, the first orally efficacious MDM2 degrader developed using PROTAC technology. MD-4251 induces potent and rapid MDM2 degradation in RS4;11 cells (DC50 = 0.2 nM; Dmax = 96% at 2 h), leading to robust p53 activation. It selectively inhibits the growth of acute leukemia cell lines with wild-type p53, with minimal activity in p53 mutant lines. MD-4251 shows excellent oral bioavailability in mice, favorable metabolic stability, and no CYP or hERG liabilities. A single oral dose induces sustained MDM2 depletion and attains complete tumor regression in vivo. These results support MD-4251 as a promising therapeutic candidate for cancers through depletion of MDM2.
{"title":"MD-4251: A First-in-Class Oral MDM2 Degrader Inducing Complete Tumor Regression with Single-Dose Administration.","authors":"Ranjan Kumar Acharyya, Liyue Huang, Angelo Aguilar, Biao Hu, Longchuan Bai, Hoda Metwally, Donna McEachern, Wei Jiang, Yu Wang, Qiuxia Li, Bo Wen, Duxin Sun, Shaomeng Wang","doi":"10.1021/acs.jmedchem.5c00809","DOIUrl":"10.1021/acs.jmedchem.5c00809","url":null,"abstract":"<p><p>MDM2 is a key negative regulator of the tumor suppressor p53 and an attractive target for cancer therapy. We report the discovery of MD-4251, the first orally efficacious MDM2 degrader developed using PROTAC technology. MD-4251 induces potent and rapid MDM2 degradation in RS4;11 cells (DC<sub>50</sub> = 0.2 nM; <i>D</i><sub>max</sub> = 96% at 2 h), leading to robust p53 activation. It selectively inhibits the growth of acute leukemia cell lines with wild-type p53, with minimal activity in p53 mutant lines. MD-4251 shows excellent oral bioavailability in mice, favorable metabolic stability, and no CYP or hERG liabilities. A single oral dose induces sustained MDM2 depletion and attains complete tumor regression in vivo. These results support MD-4251 as a promising therapeutic candidate for cancers through depletion of MDM2.</p>","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":" ","pages":"13249-13267"},"PeriodicalIF":6.8,"publicationDate":"2025-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144281699","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}
G1 to S phase transition 1 (GSPT1, also known as eRF3a) is a crucial translation termination factor that plays a vital role in acute myeloid leukemia (AML) and MYC-driven lung cancer. Degrading GSPT1 can induce apoptosis in cancer cells and reduce their viability, thus making GSPT1 a potential therapeutic target. This perspective aims to introduce the current research status of the mechanism of molecular glues targeting GSPT1, summarize the recent progress in and challenges for existing molecular glues, bifunctional degraders, and antibody-enabled molecular glues targeting GSPT1, and outline the development strategies for targeting GSPT1 in the treatment of cancer.
{"title":"Research Progress in Targeting GSPT1: Molecular Glues, Bifunctional Degraders, and Antibody-Enabled Molecular Glues for Cancer Therapy.","authors":"Binbin Cheng, Yaping Wang, Yimeng Hong, Yingxing Zhou, Jianjun Chen, Chunlai Zeng","doi":"10.1021/acs.jmedchem.5c00783","DOIUrl":"10.1021/acs.jmedchem.5c00783","url":null,"abstract":"<p><p>G1 to S phase transition 1 (GSPT1, also known as eRF3a) is a crucial translation termination factor that plays a vital role in acute myeloid leukemia (AML) and MYC-driven lung cancer. Degrading GSPT1 can induce apoptosis in cancer cells and reduce their viability, thus making GSPT1 a potential therapeutic target. This perspective aims to introduce the current research status of the mechanism of molecular glues targeting GSPT1, summarize the recent progress in and challenges for existing molecular glues, bifunctional degraders, and antibody-enabled molecular glues targeting GSPT1, and outline the development strategies for targeting GSPT1 in the treatment of cancer.</p>","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":" ","pages":"13169-13185"},"PeriodicalIF":6.8,"publicationDate":"2025-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144504173","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}
Pub Date : 2025-07-10DOI: 10.1021/acs.jmedchem.5c00883
Wei Wu, Jiaen Huang, Yuling Li, Jiaxi Chen, Xiaowei Kuang, Manzhen Ye, Rui Chen, Junli An, Zunnan Huang, Jing Sun
Cancer immunotherapy has revolutionized oncology by leveraging host immunity to eliminate malignant cells. In this study, five iridium(III) complexes (Ir1–Ir5) were synthesized and evaluated for their in vitro antitumor activity against various tumor cell lines. Among these, Ir4 and Ir5 exhibited the highest cytotoxicity and the most rapid cellular uptake in MDA-MB-231 cells. Colocalization experiments confirmed their accumulation in the endoplasmic reticulum (ER) and their ability to induce pyroptosis. Additionally, both complexes triggered ER stress, leading to increased calreticulin exposure on the cell surface, high-mobility group box 1 secretion, and ATP release, which collectively promoted immunogenic cell death. In vivo, a triple-dose Ir4 vaccine regimen significantly suppressed tumor growth compared to other treatment groups. These findings highlight the potential of Ir4-based novel triple-dose vaccination as a promising cancer immunotherapy strategy.
{"title":"Endoplasmic Reticulum-Targeting Iridium(III) Complexes Induce Pyroptosis and Enhance Immunogenic Cell Death in MDA-MB-231 Cells","authors":"Wei Wu, Jiaen Huang, Yuling Li, Jiaxi Chen, Xiaowei Kuang, Manzhen Ye, Rui Chen, Junli An, Zunnan Huang, Jing Sun","doi":"10.1021/acs.jmedchem.5c00883","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.5c00883","url":null,"abstract":"Cancer immunotherapy has revolutionized oncology by leveraging host immunity to eliminate malignant cells. In this study, five iridium(III) complexes (<b>Ir1</b>–<b>Ir5</b>) were synthesized and evaluated for their in vitro antitumor activity against various tumor cell lines. Among these, <b>Ir4</b> and <b>Ir5</b> exhibited the highest cytotoxicity and the most rapid cellular uptake in MDA-MB-231 cells. Colocalization experiments confirmed their accumulation in the endoplasmic reticulum (ER) and their ability to induce pyroptosis. Additionally, both complexes triggered ER stress, leading to increased calreticulin exposure on the cell surface, high-mobility group box 1 secretion, and ATP release, which collectively promoted immunogenic cell death. In vivo, a triple-dose <b>Ir4</b> vaccine regimen significantly suppressed tumor growth compared to other treatment groups. These findings highlight the potential of <b>Ir4</b>-based novel triple-dose vaccination as a promising cancer immunotherapy strategy.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"9 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144603972","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}