Longbin Liu, Peter D. Johnson, Matthew R. Mills, Penelope A. Turner, Michael E. Prime, Christopher J. Brown, Adrian Kotey, Samuel Coe, Paul R. Giles, Catherine Lloyd, Sarah Hayes, Frederic J. Marlin, Filippo Rota, Frank Herrmann, Manuela Heßmann, Sabine Schaertl, Franziska Zajicek, Stef De Lombaerde, Filipe Elvas, Jeroen Verhaeghe, Steven Staelens, Daniele Bertoglio, Xuemei Chen, Michael P. Conlon, Randall Davis, Samantha F. Ensor, James Haber, Kathryn C. Haber, Ming Min Hsai, John E. Mangette, William F. Penniman, Luca Anzillotti, Simone Esposito, Angelo Lembo, Laura Orsatti, Davide Ventre, Maria Veneziano, Christine Sandiego, Scott Haller, Gwendolyn A. Marriner, Ignacio Munoz-Sanjuan, Vinod Khetarpal, Jonathan A. Bard, Celia Dominguez
{"title":"Correction to “Isoindolinone-Based PET Tracers for Imaging Mutant Huntingtin Aggregates”","authors":"Longbin Liu, Peter D. Johnson, Matthew R. Mills, Penelope A. Turner, Michael E. Prime, Christopher J. Brown, Adrian Kotey, Samuel Coe, Paul R. Giles, Catherine Lloyd, Sarah Hayes, Frederic J. Marlin, Filippo Rota, Frank Herrmann, Manuela Heßmann, Sabine Schaertl, Franziska Zajicek, Stef De Lombaerde, Filipe Elvas, Jeroen Verhaeghe, Steven Staelens, Daniele Bertoglio, Xuemei Chen, Michael P. Conlon, Randall Davis, Samantha F. Ensor, James Haber, Kathryn C. Haber, Ming Min Hsai, John E. Mangette, William F. Penniman, Luca Anzillotti, Simone Esposito, Angelo Lembo, Laura Orsatti, Davide Ventre, Maria Veneziano, Christine Sandiego, Scott Haller, Gwendolyn A. Marriner, Ignacio Munoz-Sanjuan, Vinod Khetarpal, Jonathan A. Bard, Celia Dominguez","doi":"10.1021/acs.jmedchem.5c02677","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.5c02677","url":null,"abstract":"The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jmedchem.5c02677. (CSV) Correction\u0000to “Isoindolinone-Based PET Tracers\u0000for Imaging Mutant Huntingtin Aggregates” <span> 2 </span><span> views </span> <span> 0 </span><span> shares </span> <span> 0 </span><span> downloads </span> Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html. This article has not yet been cited by other publications.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"28 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145141035","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":"ZJK-807: A Selective PROTAC Degrader of KRASG12D Overcoming Resistance in Pancreatic Cancer.","authors":"Zhaojuan Liu,Heping Zheng,Yanqing Tian,Zhuoyue Li,Sai Zhang,Siqi Zhang,Shumin Ma,Xiao Wang,Chong Qin","doi":"10.1021/acs.jmedchem.5c01034","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.5c01034","url":null,"abstract":"Pancreatic cancer driven by the KRASG12D mutation faces therapeutic challenges from KRAS undruggability and acquired resistance to inhibitors like MRTX1133 via secondary mutations (e.g., Q95/Y96). Here, we report ZJK-807, a novel cereblon (CRBN)-based proteolysis-targeting chimera (PROTAC), conjugating a KRASG12D inhibitor to a CRBN ligand. It selectively degrades KRASG12D (DC50 = 79.5 ± 5.4 nM in AsPC-1 cells) with minimal impact on wild-type KRAS or other mutants (G12C/S/V, G13D), inducing mutant-specific cytotoxicity. Critically, ZJK-807 overcomes secondary mutation resistance by degrading mutant KRASG12D and suppressing resistant cell growth where MRTX1133 fails. Transcriptomic analysis revealed that ZJK-807 suppresses RAS/MAPK signaling and uniquely modulates TNF signaling and eukaryotic ribosome biogenesis, suggesting distinct mechanistic advantages. In vivo, ZJK-807 (30 mg/kg, subcutaneous) achieved 47% tumor growth inhibition in AsPC-1 xenografts with favorable pharmacokinetics. This study presents a CRBN-based PROTAC to selectively target and degrade resistant KRASG12D mutants, establishing a groundbreaking approach for KRAS-driven malignancies.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"87 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145140138","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}
Tyler G Fenske,John L McKee,Natalie G Cavalco,Serena S Schalk,Emma M Bonniwell,Josie C Lammers,Naomi Shacham,Bruna Cuccurazzu,Adam L Halberstadt,John D McCorvy
{"title":"Discovery of Highly Selective 5-HT2A Agonists Using Structure-Guided Design.","authors":"Tyler G Fenske,John L McKee,Natalie G Cavalco,Serena S Schalk,Emma M Bonniwell,Josie C Lammers,Naomi Shacham,Bruna Cuccurazzu,Adam L Halberstadt,John D McCorvy","doi":"10.1021/acs.jmedchem.5c01855","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.5c01855","url":null,"abstract":"With a resurgence in interest in psychedelics as rapid-acting and durable neuroplastic therapies, there is a critical need to develop more selective 5-HT2A agonists to investigate the basic neurobiological mechanisms of psychedelics. Here, we show that selectivity for 5-HT2A over the closely related 5-HT2C receptor can be leveraged using structure-based design to target residue L1232.53 in transmembrane 2 (TM2) of the extended binding pocket by increasing steric aliphatic bulk on the α-methylene group of the N-benzyl chemical scaffold. Furthermore, we comprehensively confirm selectivity at 5-HT2C RNA editing isoforms, TM2 reciprocal 5-HT2A and 5-HT2C mutants, and mouse 5-HT2A and 5-HT2C orthologs, to form a complete profile for highly selective 5-HT2A agonists to date. Using a combination of structure-activity relationships, molecular docking, and mouse head-twitch response assays, we show that 5-HT2A-selective agonists can be rationally designed to improve 5-HT2A target engagement, further advancing the study into the neurobiological mechanisms of psychedelic effects.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"42 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145140152","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}
Rosa De Gregorio,Federica Moraca,Pasquale Rapacciuolo,Bianca Fiorillo,Elva Morretta,Cristina Di Giorgio,Silvia Marchianò,Ginevra Lachi,Carmen Massa,Benedetta Sensini,Michele Biagioli,Lucio Spinelli,Maria Chiara Monti,Bruno Catalanotti,Valentina Sepe,Stefano Fiorucci,Angela Zampella
{"title":"Harnessing the Estradienone Scaffold to Develop Dual GPBAR1 and LIFR Modulators for Liver Fibrosis.","authors":"Rosa De Gregorio,Federica Moraca,Pasquale Rapacciuolo,Bianca Fiorillo,Elva Morretta,Cristina Di Giorgio,Silvia Marchianò,Ginevra Lachi,Carmen Massa,Benedetta Sensini,Michele Biagioli,Lucio Spinelli,Maria Chiara Monti,Bruno Catalanotti,Valentina Sepe,Stefano Fiorucci,Angela Zampella","doi":"10.1021/acs.jmedchem.5c00705","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.5c00705","url":null,"abstract":"Fibrosis is a pathological process characterized by excessive deposition of the extracellular matrix (ECM) within tissues. Chronic fibrotic disorders involving the lungs, liver, intestine, and kidneys represent a major cause of morbidity and mortality and remain a major unmet therapeutic need. In the liver, the development of pathological ECM depends on the activation of key cell targets, i.e., the hepatic stellate cells (HSC). HSCs express the leukemia inhibitory factor receptor (LIFR), which promotes fibrosis, and a bile acid-activated receptor, GPBAR1, which attenuates HSC activation. Herein, we report the design and synthesis of a new class of 4,9-estradien-3,17-dione derivatives acting as dual LIFR inhibitors and GPBAR1 agonists. In silico and pharmacological characterization of these dual modulators led to the identification of compound 2o as a first-in-class LIFR/GPBAR1 modulator that reverses liver fibrosis in vitro and in vivo. These findings demonstrate the therapeutic potential of LIFR/GPBAR1 hybrid molecules in human fibrotic disorders.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"10 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145127069","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}
Yi Xia, Yang Liu, Jinqiao Wan, Menghua Wang, Palma Rocchi, Fanqi Qu, Juan L. Iovanna, Ling Peng
{"title":"Correction to “Novel Triazole Ribonucleoside Down-Regulates Heat Shock Protein 27 and Induces Potent Anticancer Activity on Drug-Resistant Pancreatic Cancer”","authors":"Yi Xia, Yang Liu, Jinqiao Wan, Menghua Wang, Palma Rocchi, Fanqi Qu, Juan L. Iovanna, Ling Peng","doi":"10.1021/acs.jmedchem.5c02592","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.5c02592","url":null,"abstract":"In Figure 9A, tumor volumes were incorrectly calculated. The corrected Figure 9A is provided below in this Addition/Correction. This error does not affect the statistical significance of the results or the conclusions of the study. We apologize for this error. Figure 9. (A) Effect of <b>3e</b> treatment on drug-resistant MiaPaCa-2 tumor growth in vivo. Nude mice bearing MiaPaCa-2 tumors of 100 mm<sup>3</sup> were randomly selected for treatment with <b>3e</b> and no treatment as control. <b>3e</b> was injected intraperitoneally (150 mg/kg/mice) every 3 days for 5 weeks, and tumor volume was measured once a week. All of the results were expressed as the mean ± SE (<i>n</i> = 8): points are the means values of analysis; bars are the standard errors; *, differing from control (<i>p</i> ≤ 0.05), **, differing from control (<i>p</i> ≤ 0.01) by Student’s <i>t</i> test. This article has not yet been cited by other publications.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"29 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145127952","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":"PARP7: an Emerging Therapeutic Target-Insights into Biological Functions and Advances in Small-Molecule Inhibitor Development","authors":"Feihuang Qiu, Jianqiao Wang, Kehan Huang, Tianxiang Shang, Chenghao Wang, Wenxin Yan, Yungen Xu, Hongfeng Gu","doi":"10.1021/acs.jmedchem.5c01335","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.5c01335","url":null,"abstract":"PARP7 is a critically important member of the PARP family. Current research indicates that PARP7 is associated with various diseases, including viral infections, cancers, and stroke. Consequently, pharmacological modulation of PARP7 enzymatic activity has emerged as a crucial strategy for treating numerous diseases. Notably, the selective PARP7 inhibitor RBN-2397 for tumor treatment has entered Phase II trials, highlighting the translational potential of targeting this enzyme. This perspective summarizes the biological functions of PARP7 and its roles in various diseases. Furthermore, to advance the study of PARP7 inhibitors, we analyze the mechanisms of action and design strategies of existing PARP7 inhibitors as reported in current papers and patents. Additionally, we share our experiences in designing PARP7 inhibitors and present our insights regarding the opportunities and challenges in this pioneering research field.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"22 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145133558","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":"A Nitroreductase-Activated Chemiluminescent Prodrug for Real-Time Monitoring of Camptothecin Release in Peritoneal Metastasis Theranostics","authors":"Lianzhu Wei, Yumei Wu, Yanhan Zhou, Maocheng Yang, Hongyu Li, Wen-Chao Geng, Zeli Yuan, Jie Gao","doi":"10.1021/acs.jmedchem.5c01674","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.5c01674","url":null,"abstract":"While most theranostic prodrugs utilize fluorescence for reporting, chemiluminescence offers superior signal-to-noise ratios. However, chemiluminescence-based prodrug systems remain relatively scarce. Herein, we report the development of CPT-NBz-CL, a novel hypoxia-responsive theranostic prodrug activated by nitroreductase (NTR) via a self-immolative linker mechanism. This activation concomitantly releases a potent chemotherapeutic camptothecin (CPT) and generates a robust chemiluminescence signal for reporting. Encapsulation of CPT-NBz-CL into nanoparticles (CPT-NBz-CL NPs) facilitated in vivo application. In vitro studies demonstrated hypoxia-selective cytotoxicity and chemiluminescence generation. In a murine 4T1-Luc1 peritoneal metastasis model, CPT-NBz-CL NPs delineated hypoxic tumor regions through in vivo chemiluminescence imaging, reflecting the endogenous NTR activity. Crucially, CPT-NBz-CL NPs showed antitumor efficacy comparable to free CPT while significantly mitigating the parent drug’s systemic toxicity. This work validates an NTR-triggered, self-immolative chemiluminescence platform as a promising strategy for integrating high-sensitivity imaging with potent, targeted cancer therapy, offering an improved safety profile.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"82 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145133559","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":"Proteome-Wide Ligand and Target Discovery by Using Covalent Probes and Natural Products","authors":"Jingqing Liu, Chaoming Huang, Shengrong Li, Lijie Peng, Yaoliang Sun, Jianzhang Yang, Shan Li, Shilin Xu, Xiaoyun Lu, Ke Ding, Zhengqiu Li","doi":"10.1021/acs.jmedchem.5c01277","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.5c01277","url":null,"abstract":"Due to the lack of effective therapeutic targets for drug development, many diseases remain difficult to treat. To address this issue, phenotypic screening integrated with chemical proteomics has emerged as an efficient strategy to expand the scope of druggable targets. In this study, we constructed a covalent probe library based on diverse covalent kinase inhibitors and natural products containing an α, β-unsaturated ketone electrophilic warhead. Antiproliferation screening revealed that these probes exhibit potent anticancer activity against triple-negative breast cancer (TNBC) and human colon cancer. Subsequent proteomic studies identified a series of novel covalently ligandable targets, including ASNS, AKR1C1, DDX39B, and PRMT5. Functional validation demonstrated that DDX39B may represent a new therapeutic target for TNBC. Moreover, we identified a series of highly selective covalent probes targeting ARK1C1, PDIA1, and ALDH1A1, which could serve as valuable tools for detecting the expression and activity of these critical proteins.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"16 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145133637","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}
Yulong He,Shunyi Li,Yuqi Chen,Yiyang Xu,Yujie Wang,Zonglong Chen,Heyao Wang,Yingxia Li
{"title":"Discovery of Potent and Selective FABP4/5 Inhibitors with an Isoquinolone Scaffold as Potential Therapeutics for Inflammation-Related Diseases.","authors":"Yulong He,Shunyi Li,Yuqi Chen,Yiyang Xu,Yujie Wang,Zonglong Chen,Heyao Wang,Yingxia Li","doi":"10.1021/acs.jmedchem.5c01256","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.5c01256","url":null,"abstract":"Fatty acid-binding proteins 4 (FABP4) and 5 (FABP5) have emerged as promising therapeutic targets for inflammation-related diseases. Herein, we report a series of potent and selective FABP4/5 inhibitors featuring an isoquinolone scaffold through scaffold hopping of RO6806051, a dual FABP4/5 inhibitor. Among these, Y18 was identified as the most promising compound, exhibiting potent inhibitory activity with Ki values of 0.41 and 2.53 μM for FABP4 and FABP5, respectively. Notably, Y18 achieves significantly improved selectivity over FABP3 (Ki = 59.72 μM) compared to RO6806051, along with favorable pharmacokinetic properties, including high oral exposure and acceptable bioavailability. Oral administration of Y18 exhibited significant anti-inflammatory effects and attenuated LPS-induced liver injury. As an anti-inflammatory compound, Y18 demonstrates an excellent safety profile with low hERG inhibition and an LD50 value greater than 2000 mg/kg. Taken together, Y18 represents a promising dual FABP4/5 inhibitor candidate for the treatment of inflammation-related diseases.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"4 2 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145127167","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}
Cuixia Li, Yiran Wu, Wenli Wang, Lu Xu, Yan Zhou, Yang Yue, Tingting Wu, Meifang Yang, Yanli Qiu, Minhao Huang, Fangfang Zhou, Yiqing Zhou, Piliang Hao, Zhixiong Lin, Ming-Wei Wang, Suwen Zhao, Dehua Yang, Fei Xu, Houchao Tao
{"title":"Addition to “Structure-Based Ligand Discovery Targeting the Transmembrane Domain of Frizzled Receptor FZD7”","authors":"Cuixia Li, Yiran Wu, Wenli Wang, Lu Xu, Yan Zhou, Yang Yue, Tingting Wu, Meifang Yang, Yanli Qiu, Minhao Huang, Fangfang Zhou, Yiqing Zhou, Piliang Hao, Zhixiong Lin, Ming-Wei Wang, Suwen Zhao, Dehua Yang, Fei Xu, Houchao Tao","doi":"10.1021/acs.jmedchem.5c02597","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.5c02597","url":null,"abstract":"Figure A1. Evaluation of Compound <b>15</b> binding to FZD7-TMD. Chemical structures of intermediate <b>A1</b>, compound <b>15</b>, and compound <b>40</b> (A). SPR analysis of the interaction between compound <b>15</b> and FZD7-TMD (B). Molecular docking of compound <b>15</b> with FZD7 (C) and its interactions with the FZD7 binding pocket (D). Figure A2. Binding analysis of compound F7H and compound <b>28</b> using MST assay. Dose–response binding curves of compound F7H (A), compound <b>28</b> (B) with FZD7-TMD protein. Figure A3. Thermal shift assay to assess compound <b>28</b> binding to various proteins. FZD1 (A), FZD4 (B), FZD6 (C), FZD7 (D), GPR15 (E), and compounds only (F). Figure A4. TopFlash assay in FZD1–10 knockout cells. Cells were transfected with PTT5 (A), WT-FZD7 (B), or the mutant K533A (C) and then treated with IWP2 or compound <b>28</b>. Figure A5. Molecular docking results of compound <b>28</b> with firefly luciferase (PDB: 1LCI). This article references 4 other publications. This article has not yet been cited by other publications.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":"9 1","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145116620","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}