Amanda K J P F da Silva, Eucilene K de L B Marques, Lidiane M A de Lima, Widarlane A S Alves, Dayane A Gomes, Pedro L Guzzo, Mônica F Belian, Wagner E Silva, Eduardo C Lira
Bioactive compositions containing vanadium complexes have been a viable strategy for constructing more biocompatible and less toxic systems. Therefore, this work aim to develop a new composition formed by an oxidovanadium(IV) complex as levan. The acute oral toxicity and insulin resistance (IR) are investigated in an animal model using adult Swiss mice treated with daily injections of the synthetic glucocorticoid dexamethasone. The complex is characterized by electronic absorption (λmax = 771 and 880 nm) and infrared spectroscopies (3359, 3167, 1606, 1342, 1072 cm-1, and the VO at 937 cm-1); NMR of the 1H, 13C, and 51V (-427, -509, and -529 ppm), and electron paramagnetic resonance (g-factor = 1.985). The vanadium complex is classified in category 4, according to the acute toxicity protocol. IR in mice is accompanied by a rise in fasting blood glucose at seventh (2.2-fold) and 14th (threefold) days, triglyceride levels at seventh (2.6-fold) and 14th (threefold) days, and triglyceride/glucose index (TyG) at seventh (20%) and 14th (25%) days. The bioactive composition attenuated both the hyperglycemia (≈65%) and hypertriglyceridemia and TyG in a dose-dependent manner. The proposed composition shows promise in reducing IR induced by exogenous corticosteroid treatment.
{"title":"Evaluation of the Therapeutic Potential of Bioactive Materials Based on a Complex of Oxidovanadium(IV) and Exopolysaccharide Levan in a Model of Insulin Resistance in Mice.","authors":"Amanda K J P F da Silva, Eucilene K de L B Marques, Lidiane M A de Lima, Widarlane A S Alves, Dayane A Gomes, Pedro L Guzzo, Mônica F Belian, Wagner E Silva, Eduardo C Lira","doi":"10.1002/cmdc.202500754","DOIUrl":"https://doi.org/10.1002/cmdc.202500754","url":null,"abstract":"<p><p>Bioactive compositions containing vanadium complexes have been a viable strategy for constructing more biocompatible and less toxic systems. Therefore, this work aim to develop a new composition formed by an oxidovanadium(IV) complex as levan. The acute oral toxicity and insulin resistance (IR) are investigated in an animal model using adult Swiss mice treated with daily injections of the synthetic glucocorticoid dexamethasone. The complex is characterized by electronic absorption (λ<sub>max</sub> = 771 and 880 nm) and infrared spectroscopies (3359, 3167, 1606, 1342, 1072 cm<sup>-1</sup>, and the VO at 937 cm<sup>-1</sup>); NMR of the <sup>1</sup>H, <sup>13</sup>C, and <sup>51</sup>V (-427, -509, and -529 ppm), and electron paramagnetic resonance (g-factor = 1.985). The vanadium complex is classified in category 4, according to the acute toxicity protocol. IR in mice is accompanied by a rise in fasting blood glucose at seventh (2.2-fold) and 14th (threefold) days, triglyceride levels at seventh (2.6-fold) and 14th (threefold) days, and triglyceride/glucose index (TyG) at seventh (20%) and 14th (25%) days. The bioactive composition attenuated both the hyperglycemia (≈65%) and hypertriglyceridemia and TyG in a dose-dependent manner. The proposed composition shows promise in reducing IR induced by exogenous corticosteroid treatment.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202500754"},"PeriodicalIF":3.4,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145773006","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}
Anita Maksutova, Thomas M Geiger, Lorenzo Cianni, Dominika E Pieńkowska, Jan Gerhartz, Lina Read, Aleša Bricelj, Alexander Herrmann, Maurice Leon Nelles, Yuen Lam Dora Ng, Marcus D Hartmann, Jan Krönke, Izidor Sosič, Radosław P Nowak, Michael Gütschow, Christian Steinebach
A straightforward method for creating C(sp2)-C(sp3) bonds is employed to develop novel cereblon (CRBN) E3 ligase ligands, essential for targeted protein degradation (TPD) applications. While most prior studies focus on biological activities, this work explores how the linker attachment and bond types affect physicochemical stability, binding affinity, and degrading performance. Utilizing N-hydroxyphthalimide (NHP) esters and aryl bromides, a resilient decarboxylative cross-coupling technique that broadens the available chemical space beyond traditional C(sp2)-N connections is developed. Several well-established and underexplored CRBN binders and their derivatives are synthesized and studied. Binding affinity, aqueous solubility, stability in microsomes, and degradation of typical CRBN ligand off-targets are then investigated. Selected compounds are transformed into GSPT1-targeting molecular glue degraders or BRD4-targeting proteolysis-targeting chimeras (PROTACs). Benzamide-based degraders obtained using the new method have a very high ability to break down BRD4. This research shows that C(sp2)-C(sp3) connections open up new ways to fine-tune PROTAC characteristics, which unlock degrader chemotypes that were not accessible before. The results demonstrate the importance of synthetic innovation in developing ligands for TPD applications.
{"title":"A Facile Protocol for C(sp<sup>2</sup>)-C(sp<sup>3</sup>) Bond Formation Reactions Toward Functionalized E3 Ligase Ligands.","authors":"Anita Maksutova, Thomas M Geiger, Lorenzo Cianni, Dominika E Pieńkowska, Jan Gerhartz, Lina Read, Aleša Bricelj, Alexander Herrmann, Maurice Leon Nelles, Yuen Lam Dora Ng, Marcus D Hartmann, Jan Krönke, Izidor Sosič, Radosław P Nowak, Michael Gütschow, Christian Steinebach","doi":"10.1002/cmdc.202500929","DOIUrl":"https://doi.org/10.1002/cmdc.202500929","url":null,"abstract":"<p><p>A straightforward method for creating C(sp<sup>2</sup>)-C(sp<sup>3</sup>) bonds is employed to develop novel cereblon (CRBN) E3 ligase ligands, essential for targeted protein degradation (TPD) applications. While most prior studies focus on biological activities, this work explores how the linker attachment and bond types affect physicochemical stability, binding affinity, and degrading performance. Utilizing N-hydroxyphthalimide (NHP) esters and aryl bromides, a resilient decarboxylative cross-coupling technique that broadens the available chemical space beyond traditional C(sp<sup>2</sup>)-N connections is developed. Several well-established and underexplored CRBN binders and their derivatives are synthesized and studied. Binding affinity, aqueous solubility, stability in microsomes, and degradation of typical CRBN ligand off-targets are then investigated. Selected compounds are transformed into GSPT1-targeting molecular glue degraders or BRD4-targeting proteolysis-targeting chimeras (PROTACs). Benzamide-based degraders obtained using the new method have a very high ability to break down BRD4. This research shows that C(sp<sup>2</sup>)-C(sp<sup>3</sup>) connections open up new ways to fine-tune PROTAC characteristics, which unlock degrader chemotypes that were not accessible before. The results demonstrate the importance of synthetic innovation in developing ligands for TPD applications.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202500929"},"PeriodicalIF":3.4,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145766678","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}
pH-sensitive liposomes represent a promising platform for targeted drug delivery, with imidazole-based lipids offering unique advantages for tumor-specific release and enhanced endosomal escape. This pH-responsiveness facilitates cellular uptake through electrostatic interactions and improves intracellular delivery, primarily via the proton sponge effect and membrane fusion mechanisms. This article provides a comprehensive overview of imidazole-modified liposomes, detailing their pH-responsiveness mechanisms, diverse formulations, and highlighting current gaps and future research directions within the field. Special attention is given to the characteristics, advantages, and applications of key structures, including imidazole-lipid compounds, imidazole-cholesterol conjugates, and histidine-bearing polymers. The incorporation of imidazole has been shown to optimize drug release profiles, enhance intracellular delivery, and improve cytotoxicity toward cancer cells, while maintaining a favorable safety profile under physiological conditions comparable to conventional liposomes. Recent advancements highlight the impact of structural modifications, such as carbon chain length optimization, conjugated moieties, and polymer architecture, on modulating liposome stability, drug release kinetics, and targeting efficiency. Despite these notable advancements, significant challenges persist, including the critical balance between carrier stability and pH sensitivity, immune clearance, endosomal entrapment, and the complexities associated with large-scale synthesis.
{"title":"Imidazole-Based Liposomes: Emerging pH-Sensitive Drug Delivery Tools.","authors":"Faeze Shojaeinia, Mostafa Amirinejad, Atoosa Haghighizadeh, Omid Rajabi","doi":"10.1002/cmdc.202500709","DOIUrl":"https://doi.org/10.1002/cmdc.202500709","url":null,"abstract":"<p><p>pH-sensitive liposomes represent a promising platform for targeted drug delivery, with imidazole-based lipids offering unique advantages for tumor-specific release and enhanced endosomal escape. This pH-responsiveness facilitates cellular uptake through electrostatic interactions and improves intracellular delivery, primarily via the proton sponge effect and membrane fusion mechanisms. This article provides a comprehensive overview of imidazole-modified liposomes, detailing their pH-responsiveness mechanisms, diverse formulations, and highlighting current gaps and future research directions within the field. Special attention is given to the characteristics, advantages, and applications of key structures, including imidazole-lipid compounds, imidazole-cholesterol conjugates, and histidine-bearing polymers. The incorporation of imidazole has been shown to optimize drug release profiles, enhance intracellular delivery, and improve cytotoxicity toward cancer cells, while maintaining a favorable safety profile under physiological conditions comparable to conventional liposomes. Recent advancements highlight the impact of structural modifications, such as carbon chain length optimization, conjugated moieties, and polymer architecture, on modulating liposome stability, drug release kinetics, and targeting efficiency. Despite these notable advancements, significant challenges persist, including the critical balance between carrier stability and pH sensitivity, immune clearance, endosomal entrapment, and the complexities associated with large-scale synthesis.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202500709"},"PeriodicalIF":3.4,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145720083","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}
Assessing if compounds with intracellular targets reach their site of action is crucial for success in drug development. Cell type-specific uptake goes beyond permeability studies, typically mimicking crossing the gut, the lung, or the blood-brain barrier. A medium- to high-throughput cellular accumulation protocol in 96-well format is presented using six compounds, evaluating optimal conditions varying several parameters, such as incubation time, compound concentration, and extraction protocol. An optimized assay protocol for cellular accumulation of distinct chemical classes is a compromise: No one-extraction-protocol-fits-all exists; equally, some compounds need longer incubation periods to reach maximal intracellular concentration. Reliable high performance liquid chromatography with tandem mass spectrometry based quantification of cellular accumulation for all six compounds to the nM range is achieved with a short 1 h incubation. Intracellular concentrations per cell count are determined in A549ACE+TMPRSS2 cells, taking nonspecific binding into account. Hence, this approach adds valuable information during the pre-screening of compounds with intracellular targets. Finally, optimal assay conditions are emphasized as essential for predicting activity in vitro and in vivo, based on biochemical information and intracellular concentrations. In summary, the workflow for cellular accumulation determination can serve two scenarios: 1) Pre-selection of compounds for screening purposes or 2) systematic optimization of conditions to advance compounds with intracellular targets.
{"title":"A Streamlined High Performance Liquid Chromatography with Tandem Mass Spectrometry Based Workflow for Rapid Screening of Cellular Accumulation of Small Molecules.","authors":"Alina Metzen, Katharina Rox","doi":"10.1002/cmdc.202500753","DOIUrl":"https://doi.org/10.1002/cmdc.202500753","url":null,"abstract":"<p><p>Assessing if compounds with intracellular targets reach their site of action is crucial for success in drug development. Cell type-specific uptake goes beyond permeability studies, typically mimicking crossing the gut, the lung, or the blood-brain barrier. A medium- to high-throughput cellular accumulation protocol in 96-well format is presented using six compounds, evaluating optimal conditions varying several parameters, such as incubation time, compound concentration, and extraction protocol. An optimized assay protocol for cellular accumulation of distinct chemical classes is a compromise: No one-extraction-protocol-fits-all exists; equally, some compounds need longer incubation periods to reach maximal intracellular concentration. Reliable high performance liquid chromatography with tandem mass spectrometry based quantification of cellular accumulation for all six compounds to the nM range is achieved with a short 1 h incubation. Intracellular concentrations per cell count are determined in A549<sup>ACE+TMPRSS2</sup> cells, taking nonspecific binding into account. Hence, this approach adds valuable information during the pre-screening of compounds with intracellular targets. Finally, optimal assay conditions are emphasized as essential for predicting activity in vitro and in vivo, based on biochemical information and intracellular concentrations. In summary, the workflow for cellular accumulation determination can serve two scenarios: 1) Pre-selection of compounds for screening purposes or 2) systematic optimization of conditions to advance compounds with intracellular targets.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202500753"},"PeriodicalIF":3.4,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145706871","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}
Benjamin E Deprez, Debayan Dey, Natalia Zelinskaya, Erika E Csatary, Qimin Winnie Yang, Minhua Nie, Graeme L Conn, William M Wuest
One of the major mechanisms of resistance to ribosome-targeting antibiotics is the modification of ribosomal RNA (rRNA). Specific methyltransferase enzymes, for example, confer high-level resistance to aminoglycosides by selectively methylating the 16S rRNA in the ribosomal decoding center. These enzymes have been detected globally and pose a threat to the continued use of aminoglycosides. Compound 1, a dehydroamino amide inhibitor of the m1A1408 methyltransferase NpmA, was previously disclosed and identified using high-throughput virtual screening. Here, the synthesis and biological evaluation of rationally designed analogs of 1 has been reported. Guided by molecular docking, additional putative inhibitors of NpmA, as well as the functionally related m7G1405 methyltransferase RmtB, varying in each region of the original scaffold are disclosed. A modular, fragment-based synthesis enables access to 17 analogs, which exhibits mixed activity against NpmA and RmtB, including several that are selective for RmtB. The structure-activity relationship determined for the dehydroamino amide series will guide continued research against this target class with the aim of developing a toolkit for selective- or pan-16S rRNA methyltransferase inhibition.
{"title":"Design, Synthesis, and Evaluation of Novel Inhibitors of Aminoglycoside-Resistance 16S Ribosomal RNA Methyltransferases.","authors":"Benjamin E Deprez, Debayan Dey, Natalia Zelinskaya, Erika E Csatary, Qimin Winnie Yang, Minhua Nie, Graeme L Conn, William M Wuest","doi":"10.1002/cmdc.202500889","DOIUrl":"10.1002/cmdc.202500889","url":null,"abstract":"<p><p>One of the major mechanisms of resistance to ribosome-targeting antibiotics is the modification of ribosomal RNA (rRNA). Specific methyltransferase enzymes, for example, confer high-level resistance to aminoglycosides by selectively methylating the 16S rRNA in the ribosomal decoding center. These enzymes have been detected globally and pose a threat to the continued use of aminoglycosides. Compound 1, a dehydroamino amide inhibitor of the m<sup>1</sup>A1408 methyltransferase NpmA, was previously disclosed and identified using high-throughput virtual screening. Here, the synthesis and biological evaluation of rationally designed analogs of 1 has been reported. Guided by molecular docking, additional putative inhibitors of NpmA, as well as the functionally related m<sup>7</sup>G1405 methyltransferase RmtB, varying in each region of the original scaffold are disclosed. A modular, fragment-based synthesis enables access to 17 analogs, which exhibits mixed activity against NpmA and RmtB, including several that are selective for RmtB. The structure-activity relationship determined for the dehydroamino amide series will guide continued research against this target class with the aim of developing a toolkit for selective- or pan-16S rRNA methyltransferase inhibition.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202500889"},"PeriodicalIF":3.4,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12698113/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145706847","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}
Gareth Arnott, Jonah Ruskin, Antonin Knirsch, Aditya Vardhan, Adam Huczyński, Travis Dudding, Thomas Lectka
Imagine a divalent metal ion (such as Zn(II)) binding to a folded ionophore such as salinomycin. The resulting complex is superacidified, shooting a proton at an unbound salinomycin molecule and cleaving it into two parts. However, when potassium ion is prebound to salinomycin, no superacidic protons are generated by this complex, whereas the protons generated by the divalent metal ion complex just bounce off. More details can be found in the Research Article by Adam Huczyński, Travis Dudding, Thomas Lectka, and co-workers (DOI: 10.1002/cmdc.202500783).