ChemMedChemPub Date : 2025-06-28DOI: 10.1002/cmdc.202500248
Yuequan Wu, Meng He, Zhi-Hao Li, Hongtao Kong, Ruirui Li, Ye Qu, En Zhang
{"title":"Recent Progress of Antibacterial Carbon Dots Prepared from Marketed Small Molecule Antibacterial Drugs.","authors":"Yuequan Wu, Meng He, Zhi-Hao Li, Hongtao Kong, Ruirui Li, Ye Qu, En Zhang","doi":"10.1002/cmdc.202500248","DOIUrl":"https://doi.org/10.1002/cmdc.202500248","url":null,"abstract":"<p><p>The massive consumption of antibiotics leads to antibiotic resistance, which is a major concern in the global health crisis. Therefore, the development of non-antibiotic antibacterial drugs occurs simultaneously with the development of antibiotics. Nanomaterial applications in antibacterial research are currently expanding. Carbon dots, termed as CD, are a novel class of nanomaterials that have garnered a lot of interest. Antibacterial CD has many outstanding characteristics, such as high biocompatibility, easy surface modification, excellent optical properties, an extensive precursor source, and a small particle size (1-10 nm). Although there are many potential sources of CD, many researchers find it challenging to screen precursors for the purpose of preparing CD with remarkable antibacterial abilities. It is often recommended to generate CD directly from antibacterial agents since the process usually maintains the part qualities of its precursors during preparation. In this paper, CDs prepared by marketed small molecule antibacterial drugs were selected as our topic. Their characteristics of preparation, biocompatibility and antibacterial activity were discussed. This review summarizes the antibacterial CDs prepared from aminoglycosides, β-lactams, quinolones, antituberculosis drugs, and nitroimidazole antibacterial drugs as precursors. Then the advantages and mechanisms of CDs, the existing problems and future possibilities of antibacterial CDs were also explored.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202500248"},"PeriodicalIF":3.6,"publicationDate":"2025-06-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144525690","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ChemMedChemPub Date : 2025-06-27DOI: 10.1002/cmdc.202500397
Ja Young Cho, Sanghwa Park, Taejung Kim, Junghye Eom, Jung-Rae Rho, Hyoung-Woo Bai
{"title":"Discovery of a Phenylalanine-Derived Natural Compound as a Potential Dual Inhibitor of MDM2 and MDMX.","authors":"Ja Young Cho, Sanghwa Park, Taejung Kim, Junghye Eom, Jung-Rae Rho, Hyoung-Woo Bai","doi":"10.1002/cmdc.202500397","DOIUrl":"10.1002/cmdc.202500397","url":null,"abstract":"<p><p>Dual inhibition of the negative p53 regulators MDM2 and MDMX has emerged as an effective strategy in p53-based anticancer therapy. However, dual inhibitors are limited, and many inhibitors exhibit poor pharmacokinetic properties and fast dissociation kinetics. Among newly identified microbial metabolites, the novel phenylalanine-derived compound P5 isolated from Micromonospora sp. MS-62 (FBCC-B8445) exhibits inhibitory activity against both MDM2 and MDMX. The binding of P5 to MDM2 and MDMX is demonstrated by surface plasmon resonance, which reveals nanomolar-level affinity and slow dissociation kinetics (KD = 46 nM for MDM2; 576 nM for MDMX). This dual inhibitory activity was further supported by molecular docking, which reveals binding of P5 to the p53-binding pockets of both MDM2 and MDMX through extensive noncovalent interactions. In cell-based assays, P5 reduced cancer cell viability across several human cell lines. Furthermore, in silico analysis indicates favorable pharmacokinetic properties, including gastrointestinal absorption, blood-brain barrier permeability, and compliance with Lipinski's and Veber's criteria. P5 combines dual-target engagement with binding persistence and favorable pharmacokinetic characteristics, addressing limitations of earlier inhibitors. P5 is a potential lead compound for the development of MDM2/MDMX-targeted anticancer agents.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e2500397"},"PeriodicalIF":3.6,"publicationDate":"2025-06-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144504288","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Design, Synthesis, Antimicrobial, and Antimalarial Evaluation of Quinoline Hydrazone Derivatives: Insight through Density Functional Theory Analysis, Molecular Docking, and Absorption, Distribution, Metabolism, Excretion, and Toxicity Predictions.","authors":"Sangeeta Verma, Sukhbir Lal, Rakesh Narang, Ritu Badal, Raman Saini, Somdutt Mujwar","doi":"10.1002/cmdc.202500283","DOIUrl":"10.1002/cmdc.202500283","url":null,"abstract":"<p><p>Antimicrobial and antimalarial resistance are the major global health threats and indicating to develop novel targeted molecules to overcome the resistance problem. In this concern, in the present study, some quinoline hydrazone derivatives (6a-6o) have been synthesized and evaluated as antimicrobial and antimalarial agents. Compound 6o is observed to be the most active and presented equipotent antibacterial activity (MIC = 6.25 μg mL<sup>-1</sup>) as the standard drug ofloxacin against Escherichia coli and Pseudomonas aeruginosa. Antimalarial activity showed that compound 6g exhibits maximum inhibitory action (IC<sub>50</sub> = 0.56 μg mL<sup>-1</sup>) and is significant in contrast to standard drugs chloroquine (IC<sub>50</sub> = 0.020 μg mL<sup>-1</sup>) and quinine (IC<sub>50</sub> = 0.268 μg mL<sup>-1</sup>) against Plasmodium falciparum. Density functional theory analysis evaluates the chemical reactivity of molecular orbitals of the most active compounds 6g and 6o. Furthermore, molecular docking study showed that most active antimalarial (6g) and antimicrobial (6o) compounds exhibit potent interactions with targeted enzymes (lactate dehydrogenase and dihydrofolate reductase (DHFR), respectively). In silico drug-likeness parameters and absorption, distribution, metabolism, excretion, and toxicity study indicate that all the synthesized derivatives followed the acceptance criteria. The present study emphasizes that compounds 6g (antimalarial) and 6o (antimicrobial) can be developed as novel inhibitors against lactate dehydrogenase and DHFR enzyme, respectively, on the completion of drug discovery approaches.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e2500283"},"PeriodicalIF":3.6,"publicationDate":"2025-06-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144493268","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ChemMedChemPub Date : 2025-06-26DOI: 10.1002/cmdc.202500260
Valdas Vainauskas, Povilas Kavaliauskas, Birutė Grybaitė, Vidmantas Petraitis, Rūta Petraitienė, Ramunė Grigalevičiūtė, Rūta Prakapaitė, Waldo Acevedo, Vytautas Mickevičius
{"title":"Synthesis and Characterization of Sulfonamide-Imidazole Hybrids with In Vitro Antiproliferative Activity against Anthracycline-Sensitive and Resistant H69 Small Cell Lung Cancer Cells.","authors":"Valdas Vainauskas, Povilas Kavaliauskas, Birutė Grybaitė, Vidmantas Petraitis, Rūta Petraitienė, Ramunė Grigalevičiūtė, Rūta Prakapaitė, Waldo Acevedo, Vytautas Mickevičius","doi":"10.1002/cmdc.202500260","DOIUrl":"10.1002/cmdc.202500260","url":null,"abstract":"<p><p>A series of novel sulfonamide-imidazole hybrid derivatives are synthesized, and their antiproliferative properties are evaluated. The global challenge of cancer, highlighted by rising morbidity and mortality rates, is further intensified by the increasing prevalence of drug-resistant cancer cells. Targeting the molecular mechanisms underlying therapeutic resistance is crucial for the development of innovative treatment strategies to improve clinical outcomes. Herein, the in vitro antiproliferative activity of novel sulfonamide derivatives, which exhibited significant low micromolar cytotoxicity against H69 human lung carcinoma cells and anthracycline-resistant H69AR cells compared to untreated controls (p < 0.05), is synthesized and characterized. The most promising compounds (11e, 11g, 11h, 12) also demonstrate cytotoxic activity against A549 human lung adenocarcinoma cells. Molecular docking studies predict that compound 11e interacts with tropomyosin receptor kinase A (TRKA) and mesenchymal-epithelial transition factor (c-MET) at conserved binding sites also targeted by FDA-approved inhibitors. These findings suggest that the novel sulfonamide derivatives, particularly compound 11e, may serve as promising antiproliferative candidates targeting TRKA and c-MET, potentially contributing to strategies aimed at overcoming drug resistance. Moreover, compound 11e can serve as a structural scaffold for future hit-to-lead optimization efforts.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e2500260"},"PeriodicalIF":3.6,"publicationDate":"2025-06-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144493269","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ChemMedChemPub Date : 2025-06-24DOI: 10.1002/cmdc.202401040
Federica Barbugian, Domenico Salerno, Elisa Ballarini, Luca Crippa, Oscar Francesconi, Francesco Mantegazza, Guido Cavaletti, Stefano Roelens, Gemma Leone, Simone Pepi, Luigi Talarico, Agnese Magnani, Cristina Nativi, Laura Russo
{"title":"Bioresponsive Hyaluronic Acid-Based Hydrogel Inhibits Matrix Metalloproteinase-2 in Glioblastoma Microenvironment.","authors":"Federica Barbugian, Domenico Salerno, Elisa Ballarini, Luca Crippa, Oscar Francesconi, Francesco Mantegazza, Guido Cavaletti, Stefano Roelens, Gemma Leone, Simone Pepi, Luigi Talarico, Agnese Magnani, Cristina Nativi, Laura Russo","doi":"10.1002/cmdc.202401040","DOIUrl":"https://doi.org/10.1002/cmdc.202401040","url":null,"abstract":"<p><p>Glioblastoma multiforme (GBM) is an extremely malignant cancer, resistant to standard therapies. Tumor resection is associated with better outcomes but a complete resection of GBMs is challenging for anatomical limitation and for the high degree of invasiveness. A bioresponsive hydrogel based on hyaluronic acid (HA) cross-linked with a branched metalloproteinase (MMP) inhibitor (MMPI) is proposed. The fully characterized hydrogel, HA-MMPI, presents physical properties suitable for filling the surgical resection cavity and for in situ delivery of the inhibitor. The bioresponsive material selectively inhibits MMP-2 versus MMP-9 in glioblastoma microenvironment.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e2401040"},"PeriodicalIF":3.6,"publicationDate":"2025-06-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144482694","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ChemMedChemPub Date : 2025-06-24DOI: 10.1002/cmdc.202500185
Matteo Lusardi, Elva Morretta, Andrea Spallarossa, Maria Chiara Monti, Camillo Rosano, Erika Iervasi, Marco Ponassi, Matteo Mori, Fiorella Meneghetti, Chiara Brullo
{"title":"Targeting Ubiquitin-Specific Protease 7 with Novel 5-Amino-Pyrazole Inhibitors: Design, Synthesis, and Biological Evaluation.","authors":"Matteo Lusardi, Elva Morretta, Andrea Spallarossa, Maria Chiara Monti, Camillo Rosano, Erika Iervasi, Marco Ponassi, Matteo Mori, Fiorella Meneghetti, Chiara Brullo","doi":"10.1002/cmdc.202500185","DOIUrl":"https://doi.org/10.1002/cmdc.202500185","url":null,"abstract":"<p><p>To further extend the structure-activity relationships (SARs) of the previously published ubiquitin-specific protease 7 (USP-7) inhibitor STIRUR-41, a small library of 5-aminopyrazoles 1a-d and 2a-d is designed and synthesized. The chemical identity of the desired structure is confirmed by nuclear magnetic resonance and single crystal X-ray diffraction analyses. All novel derivatives are tested as potential USP-7 inhibitors and compounds 1a-d block enzyme activity in a dose-dependent manner and with lower IC<sub>50</sub> values compared to the lead compound STIRUR-41. Notably, 1d, bearing a meta-trifluoromethylphenyl group linked to the carbamate moiety, proved to be the most active candidate. Conversely, compounds belonging to series 2, which possess greater steric hindrance, exhibit no activity. The most effective compounds of series 1 are noncytotoxic across a panel of tumor and normal cell lines at 10 μM concentration. For the most active compound 1d, a parallel artificial membrane permeability assay is also performed, as well as docking and molecular dynamics simulations.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e2500185"},"PeriodicalIF":3.6,"publicationDate":"2025-06-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144482695","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ChemMedChemPub Date : 2025-06-19DOI: 10.1002/cmdc.202500405
Claudia Finamore, Carmen Festa, Rosa Barbato, Stefano Fiorucci, Angela Zampella, Simona De Marino
{"title":"The Role of Five-Membered Aromatic Rings Containing N and O in Modulating Bile Acid Receptors: An Overview.","authors":"Claudia Finamore, Carmen Festa, Rosa Barbato, Stefano Fiorucci, Angela Zampella, Simona De Marino","doi":"10.1002/cmdc.202500405","DOIUrl":"10.1002/cmdc.202500405","url":null,"abstract":"<p><p>Over the past decades, extensive scientific research in the fields of chemistry and pharmaceutical chemistry has led to the synthesis and study of numerous chemical compounds with diverse therapeutic applications. Many of these compounds feature heterocyclic aromatic structures, including four-, five-, and six-membered rings. Among them, five-membered heteroaromatic rings have garnered particular attention in medicinal chemistry due to their favorable properties, such as enhanced metabolic stability, solubility, and bioavailability, key attributes for the development of effective drugs. The distinctive physicochemical properties and biological activities of five-membered heterocycles have established them as vital structural motifs in numerous clinically effective drugs. These heterocyclic compounds play a crucial role in the design of therapeutic agents, including those targeting bile acid receptors. Bile acid receptor modulators, activated by endogenous bile acids, offer promising potential in treating a variety of metabolic and enterohepatic disorders, such as dyslipidemia, diabetes, cholestasis, and inflammatory bowel disease. This review aims to provide an up-to-date overview of aromatic five-membered nitrogen- and oxygen-containing heterocycles, focusing on their role as bile acid receptor modulators, particularly farnesoid X receptor and G protein-coupled bile acid receptor 1. These receptors are clinically validated targets for the treatment of metabolic disorders and nonalcoholic steatohepatitis.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e2500405"},"PeriodicalIF":3.6,"publicationDate":"2025-06-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144323963","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ChemMedChemPub Date : 2025-06-19DOI: 10.1002/cmdc.202500258
Alexander Dömling, Tad A. Holak
{"title":"Balinatunfib: A Clinical Oral Small Molecule TNFα Inhibitor","authors":"Alexander Dömling, Tad A. Holak","doi":"10.1002/cmdc.202500258","DOIUrl":"10.1002/cmdc.202500258","url":null,"abstract":"<p>Most diseases are accompanied by an inflammatory response, making effective pharmacological control highly desirable. Tumor necrosis factor alpha (TNFα) is a key cytokine driving inflammatory and autoimmune diseases, such as rheumatoid arthritis and inflammatory bowel disease. Although biological TNFα inhibitors revolutionized treatment, they have drawbacks including lacking blood–brain barrier penetration, parenteral administration, and immunogenicity. Recent studies highlight the potential of small-molecule approaches to target TNFα by stabilizing an asymmetrical, receptor-incompetent trimer conformation. Balinatunfib (also known as SAR441566) is an orally available small molecule designed to exploit this mechanism, thereby preventing TNFα from effectively binding to its receptors. In preclinical models, balinatunfib reduces inflammation comparably to biologic therapies, yet avoids the complexities of large protein therapeutics. This allosteric strategy involves capturing a sampled but distorted state of TNFα, thereby blocking receptor clustering and downstream proinflammatory signaling. The oral route of administration confers practical advantages in terms of patient compliance and could facilitate drug access to sites traditionally less amenable to biologics, such as the central nervous system. By demonstrating that small molecules can achieve high-affinity, conformation-based inhibition of TNFα, balinatunfib, and related compounds may result in a new area of orally administered therapies that advance the management of TNFα-mediated diseases.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":"20 14","pages":""},"PeriodicalIF":3.6,"publicationDate":"2025-06-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cmdc.202500258","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144332194","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ChemMedChemPub Date : 2025-06-18DOI: 10.1002/cmdc.202500244
Helge Vatheuer, Jonas Paulus, Lisa Johannknecht, Gerald Keller, Rebecca Maria Ziora, Lukas Stelzl, Paul Czodrowski
{"title":"Proton first: rationalizing a proton transfer in a protein-fragment complex.","authors":"Helge Vatheuer, Jonas Paulus, Lisa Johannknecht, Gerald Keller, Rebecca Maria Ziora, Lukas Stelzl, Paul Czodrowski","doi":"10.1002/cmdc.202500244","DOIUrl":"https://doi.org/10.1002/cmdc.202500244","url":null,"abstract":"<p><p>We present a combination of experimental and theoretical approaches to decipher the molecular recognition event of benzoic acid complexed with Protein Kinase A. The publicly known crystal structure suggests the protonated form of benzoic acid to be complexed with Protein Kinase A. Such a protonation pattern of is unlikely for benzoic acid in aqueous enviromnent and must be induced by complexation to Protein Kinase A. Unfortunately, isothermal titration calorimetry does not reveal any binding event which might be caused by the low affinity. However, Poisson-Boltzmann calculations and molecular dynamics simulations strengthen the initial hypothesis of a protonated benzoic acid binding to Protein Kinase A.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202500244"},"PeriodicalIF":3.6,"publicationDate":"2025-06-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144323962","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
ChemMedChemPub Date : 2025-06-17DOI: 10.1002/cmdc.202581201
{"title":"Front Cover: (ChemMedChem 12/2025)","authors":"","doi":"10.1002/cmdc.202581201","DOIUrl":"https://doi.org/10.1002/cmdc.202581201","url":null,"abstract":"<p>Bringing chemistry, biology, and drug discovery together fosters innovation, increases collaboration, and accelerates science. <i>ChemMedChem</i> publishes high-impact articles showcasing the breadth of international research in medicinal chemistry, from small pharmacologically active molecules to new modalities including nanomedicine and biologics. Cover image provided courtesy of Ivan Sanchis and Dr. Álvaro Siano.\u0000 <figure>\u0000 <div><picture>\u0000 <source></source></picture><p></p>\u0000 </div>\u0000 </figure></p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":"20 12","pages":""},"PeriodicalIF":3.6,"publicationDate":"2025-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cmdc.202581201","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144308928","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}