Jing Pang, Shun Feng, Bin Huang, Jujun Zhou, Linjun Zhan, Ya-Qiu Long
{"title":"Novel Bioorthogonal Theranostic Scaffold Enables on-Target Drug Release and Real Time Monitoring In Vivo.","authors":"Jing Pang, Shun Feng, Bin Huang, Jujun Zhou, Linjun Zhan, Ya-Qiu Long","doi":"10.1021/acs.jmedchem.4c02965","DOIUrl":"10.1021/acs.jmedchem.4c02965","url":null,"abstract":"<p><p>Bioorthogonal chemistry-based prodrug strategy features spatiotemporally controlled release of therapeutic agent and/or imaging probe. However, the integration of diagnosis and therapy into a single molecule paired with a single bioorthogonal trigger remains a challenge. In this study, we devised a novel bioorthogonal theranostic scaffold amenable to the conjugation of various targeting agent and click-to-release reaction with the bioorthogonal prodrug to enable targeted drug liberation with concomitant fluorescence emission. Such one-stone-three-birds scaffold consists of a new fluorophore phenanthrodioxine (PDO) linked with a fluorescence masking group, tetrazine (Tz) which serves as a dual switch for the activation of fluorophore and drug. Further installation of a warhead of phenylboronic acid (PBA) ensures the targeted accumulation of the resultant PBA-PDO-Tz conjugate in tumor cells, thereby achieving on-demand activation of <i>trans</i>-cyclooctene-caged anticancer drug Doxorubicin with real-time monitoring and on-target cytotoxicity in live cells and an A549 xenograft mouse model. The targeted single trigger-dual response scaffold holds promise for precise theranostics applications <i>in vivo</i>.</p>","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":" ","pages":"3824-3836"},"PeriodicalIF":6.8,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143062268","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}
{"title":"Quantitation and Error Measurements in Dose-Response Curves.","authors":"Bharath Srinivasan, Matthew D Lloyd","doi":"10.1021/acs.jmedchem.5c00131","DOIUrl":"10.1021/acs.jmedchem.5c00131","url":null,"abstract":"","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":" ","pages":"2052-2056"},"PeriodicalIF":6.8,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143062300","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}
{"title":"Correction to “Design, Synthesis, and Structure–Activity Relationship Study of Novel Indole-2-carboxamide Derivatives as Anti-inflammatory Agents for the Treatment of Sepsis”","authors":"Zhiguo Liu, Longguang Tang, Heping Zhu, Tingting Xu, Chenyu Qiu, Suqing Zheng, Yugui Gu, Jianpeng Feng, Yali Zhang, Guang Liang","doi":"10.1021/acs.jmedchem.5c00203","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.5c00203","url":null,"abstract":"In Figure 4A, there is a typographical error in the Sham-Vehicle image due to a mistake during figure preparation. The corrected figure is provided here. Figure 4. <b>14f</b> and <b>14g</b> inhibited macrophages in filtration through F4/80-staining. (A) F4/80 immunohistochemical staining. (B) Histogram of F4/80-positive cells in A. ns: no signifance. *<i>p</i> < 0.05, **<i>p</i> < 0.01, ***<i>p</i> < 0.001 vs LPS vehicle. We have now corrected the original Figure 4, and all authors have concurred with the changes. We sincerely apologize for any confusion this may have caused and appreciate your understanding as we undertake these necessary corrections to uphold the integrity of our research findings. This article has not yet been cited by other publications.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"21 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143401883","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}
{"title":"Design of Selective BRD4 Inhibitors for the Treatment of Autosomal Dominant Polycystic Kidney Disease","authors":"Yueyue Yang, Hongli Liu, Haoxing Yuan, Kaikai Lyu, Haiyang Zhong, Yanlian Li, Danyan Cao, Wenchao Zhao, Haoran Zhang, Bing Xiong, Danqi Chen, Dong Guo","doi":"10.1021/acs.jmedchem.4c02128","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c02128","url":null,"abstract":"Epigenetic modulation plays a pivotal role in restraining tumor progression by governing gene expression and protein function. Autosomal dominant polycystic kidney disease (ADPKD), characterized by neoplastic-like progression, can be managed by inhibiting cyst expansion. Of note, the epigenetic regulator BRD4 has been implicated in ADPKD’s development. Our prior research unveiled a class of (pyrazol-3-yl) pyrimidin-4-amine compounds as potent BRD4 inhibitors with additional kinase inhibition, which might induce unwanted biological activities. To address this, this study focused on creating selective BRD4 inhibitors through structure-guided design, minimizing off-target kinase interactions. Specifically, compound <b>23</b> emerged as an efficacious and selective BRD4 inhibitor in cellular and embryonic kidney models of ADPKD, along with encouraging outcomes in murine models. Collectively, these results highlight the therapeutic potential of targeted BRD4 inhibition as a safe and efficacious strategy for managing ADPKD.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"16 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143401887","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}
Soo Bin Park, Na Young Lee, Eun-Young Lee, Subin Kim, Narasaem Lee, Eun Chae Roh, Yoon Gyoon Kim, Hee Jin Kim, Mi Sun Jin, Chul-Seung Park, Yong-Chul Kim
{"title":"Discovery of Diphenyl Ether Derivatives as Novel BK<sub>Ca</sub> Channel Activators: Structure-Activity Relationship, Cryo-EM Complex Structures, and <i>In Vivo</i> Animal Studies.","authors":"Soo Bin Park, Na Young Lee, Eun-Young Lee, Subin Kim, Narasaem Lee, Eun Chae Roh, Yoon Gyoon Kim, Hee Jin Kim, Mi Sun Jin, Chul-Seung Park, Yong-Chul Kim","doi":"10.1021/acs.jmedchem.4c02008","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c02008","url":null,"abstract":"<p><p>The BK<sub>Ca</sub> channel, a large-conductance calcium-activated potassium channel, plays a crucial role in maintaining the homeostasis of the micturition cycle and airway-related functions. In this study, we optimized a novel BK<sub>Ca</sub> channel activator, <b>4d</b>, with a diphenyl ether structure identified from library screening. This led to the discovery of potent activators, <b>10b</b> (EC<sub>50</sub> = 0.12 μM, cell-based assay) and <b>51b</b>, an orally bioavailable derivative. Compound <b>10b</b> demonstrated potent <i>in vivo</i> efficacy in a spontaneous hypertensive rat (SHR) of urinary incontinence model, while compound <b>51b</b> showed dose-dependent cough suppression efficacy with an ED<sub>50</sub> of 11.8 mg/kg in a citric acid-induced cough model. Furthermore, we reported the cryo-electron microscopy (cryo-EM) structures of the BK<sub>Ca</sub> channel in complex with <b>10b</b> and <b>51b</b> at resolutions of 2.8 and 3.4 Å. Based on structural analyses, we determined the binding sites and key interaction residues of <b>51b</b>, which were validated via mutation studies.</p>","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":" ","pages":""},"PeriodicalIF":6.8,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143412364","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}
Kevin J Filipski, Luis A Martinez-Alsina, Matthew R Reese, Edelweiss Evrard, Leanne M Buzon, Kimberly O Cameron, Yuan Zhang, Karen J Coffman, James Bradow, Bethany L Kormos, Shenping Liu, John D Knafels, Parag V Sahasrabudhe, Jie Chen, Amit S Kalgutkar, Andrew J Bessire, Christine C Orozco, Amanda Balesano, Matthew A Cerny, Eliza Bollinger, Allan R Reyes, Brigitte Laforest, Amy Rosado, George Williams, Mackenzie Marshall, Kelly Tam Neale, Xian Chen, Dinesh Hirenallur-Shanthappa, John C Stansfield, John Groarke, Ruolun Qiu, Spinel Karas, Rachel J Roth Flach, William P Esler
{"title":"Discovery of First Branched-Chain Ketoacid Dehydrogenase Kinase (BDK) Inhibitor Clinical Candidate PF-07328948.","authors":"Kevin J Filipski, Luis A Martinez-Alsina, Matthew R Reese, Edelweiss Evrard, Leanne M Buzon, Kimberly O Cameron, Yuan Zhang, Karen J Coffman, James Bradow, Bethany L Kormos, Shenping Liu, John D Knafels, Parag V Sahasrabudhe, Jie Chen, Amit S Kalgutkar, Andrew J Bessire, Christine C Orozco, Amanda Balesano, Matthew A Cerny, Eliza Bollinger, Allan R Reyes, Brigitte Laforest, Amy Rosado, George Williams, Mackenzie Marshall, Kelly Tam Neale, Xian Chen, Dinesh Hirenallur-Shanthappa, John C Stansfield, John Groarke, Ruolun Qiu, Spinel Karas, Rachel J Roth Flach, William P Esler","doi":"10.1021/acs.jmedchem.4c02230","DOIUrl":"10.1021/acs.jmedchem.4c02230","url":null,"abstract":"<p><p>Inhibition of branched-chain ketoacid dehydrogenase kinase (BDK or BCKDK), a negative regulator of branched-chain amino acid (BCAA) metabolism, is hypothesized to treat cardio-metabolic diseases. From a starting point with potential idiosyncratic toxicity risk, modification to a benzothiophene core and discovery of a cryptic pocket allowed for improved potency with 3-aryl substitution to arrive at PF-07328948, which was largely devoid of protein covalent binding liability. This BDK inhibitor was shown also to be a BDK degrader in cells and in vivo rodent studies. Plasma biomarkers, including BCAAs and branched-chain ketoacids (BCKAs), were lowered in vivo with enhanced pharmacodynamic effect upon chronic dosing due to BDK degradation. This molecule improves metabolic and heart failure end points in rodent models. PF-07328948 is the first known selective BDK inhibitor candidate to be examined in clinical studies, with Phase 1 single ascending dose data showing good tolerability and a pharmacokinetic profile commensurate with once-daily dosing.</p>","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":" ","pages":"2466-2482"},"PeriodicalIF":6.8,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142666220","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}
Paige J Monsen, Prashant V Bommi, Arabela A Grigorescu, Kristen L Lauing, Yingyu Mao, Payton Berardi, Lijie Zhai, Oluwatomilayo Ojo, Manon Penco-Campillo, Taylor Koch, Michael Egozi, Sonam Jha, Sara F Dunne, Hong Jiang, Guiqin Song, Fang Zhang, Steven Kregel, Ali Vaziri-Gohar, Sean W Fanning, Pilar Sanchez-Gomez, Jacob M Allen, Bakhtiar Yamini, Rimas V Lukas, Derek A Wainwright, Gary E Schiltz
{"title":"Rational Design and Optimization of a Potent IDO1 Proteolysis Targeting Chimera (PROTAC).","authors":"Paige J Monsen, Prashant V Bommi, Arabela A Grigorescu, Kristen L Lauing, Yingyu Mao, Payton Berardi, Lijie Zhai, Oluwatomilayo Ojo, Manon Penco-Campillo, Taylor Koch, Michael Egozi, Sonam Jha, Sara F Dunne, Hong Jiang, Guiqin Song, Fang Zhang, Steven Kregel, Ali Vaziri-Gohar, Sean W Fanning, Pilar Sanchez-Gomez, Jacob M Allen, Bakhtiar Yamini, Rimas V Lukas, Derek A Wainwright, Gary E Schiltz","doi":"10.1021/acs.jmedchem.5c00026","DOIUrl":"10.1021/acs.jmedchem.5c00026","url":null,"abstract":"<p><p>Indoleamine 2,3-dioxygenase 1 (IDO1) is an immunosuppressive protein that inhibits antitumor immunity through both tryptophan metabolism and nonenzymatic functions. Drugs targeting IDO1 enzyme activity have failed to improve the overall survival of patients with cancer. Developing new therapeutics that neutralize both enzyme- and nonenzyme-derived immunosuppressive IDO1 effects is therefore of high interest. We previously described a novel proteolysis targeting chimera (PROTAC), NU223612, that degrades IDO1 in cultured human glioblastoma (GBM) cells, as well as in well-established brain tumors, <i>in vivo</i>. In this study, we rationally optimized the structure of our lead series to create NU227326, which degrades IDO1 with a DC<sub>50</sub> of 5 nM in human GBM cells. Mechanistic studies showed that IDO1 degradation occurred through the ubiquitin-proteasome system and was sustained for at least 2 days, supporting NU227326 as a highly potent IDO1 PROTAC suitable for further studies in GBM and other human cancers.</p>","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":" ","pages":""},"PeriodicalIF":6.8,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143412368","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}
{"title":"Lactoferrin Nanoparticle-Vanadium Complex: A Promising High-Efficiency Agent against Glioblastoma by Triggering Autophagy and Ferroptosis","authors":"Shuangshuang Gai, Qiwei Yan, Shan Li, Xuwei Zhong, Yiming Qin, Ming Jiang","doi":"10.1021/acs.jmedchem.4c02696","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c02696","url":null,"abstract":"Glioblastoma represents the most aggressive type of brain cancer with minimal clinical advancements in recent decades attributed to the absence of efficient drug delivery strategies. In this study, we synthesized a series of vanadium complexes (V1–V4) and then constructed a lactoferrin (LF)-V4 nanoparticle (NP) delivery system. The nanoplatform crossed the blood-brain barrier by binding to low-density lipoprotein receptor-associated protein-1 and selectively targeted glioblastoma, ultimately inhibiting the growth of in situ glioblastoma tumors. LF-V4 NPs induced autophagic cell death in U87-MG cells by generating reactive oxygen species (ROS) that damaged the mitochondria. Further studies revealed that LF-V4 NPs triggered lipid peroxidation through the accumulation of ROS, the depletion of GSH, and the downregulation of GPX4 and SLC7A11, ultimately leading to ferroptosis in glioblastoma cells.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"2 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143401881","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}
Yang Li, Yun Liu, Dan Zhang, Juncheng Chen, Gaoxia Yang, Pan Tang, Chengcan Yang, Jie Liu, Jifa Zhang, Liang Ouyang
{"title":"Correction to \"Discovery, Synthesis, and Evaluation of Novel Dual Inhibitors of a Vascular Endothelial Growth Factor Receptor and Poly(ADP-Ribose) Polymerase for BRCA Wild-Type Breast Cancer Therapy\".","authors":"Yang Li, Yun Liu, Dan Zhang, Juncheng Chen, Gaoxia Yang, Pan Tang, Chengcan Yang, Jie Liu, Jifa Zhang, Liang Ouyang","doi":"10.1021/acs.jmedchem.5c00227","DOIUrl":"10.1021/acs.jmedchem.5c00227","url":null,"abstract":"","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":" ","pages":"3906-3907"},"PeriodicalIF":6.8,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143062161","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}