Jiri Ledvinka, Francesco Rota Sperti, Gabor Paragi, Marc Pirrotta, Nicolas Chéron, Ibai E Valverde, Petra Menova, David Monchaud
Fluorescence detection of DNA and RNA G-quadruplexes (G4s) is a very efficient strategy to assess not only the existence and prevalence of cellular G4s but also their relevance as targets for therapeutic interventions. Among the fluorophores used to this end, turn-on probes are the most interesting since their fluorescence is triggered only upon interaction with their G4 targets, which ensures a high sensitivity and selectivity of detection. We reported on a series of twice-as-smart G4 probes, which are both smart G4 ligands (whose structure is reorganized upon interaction with G4s) and smart fluorescent probes (whose fluorescence is turned on upon interaction with G4s). The fine mechanistic details behind the excellent properties of the best prototype N-TASQ remain to be deciphered: to investigate this, we report here on the synthesis and studies of two analogues, TzN-TASQ and AlkN-TASQ, and on a careful analysis of their G4-interacting properties, investigated both in vitro and in silico. Our results show that fine-tuning their constitutive structural elements allows for increasing the efficiency of both their 'off' (i. e., a conformation with a low fluorescence) and 'on' states (i. e., a conformation with a high fluorescence), which opens interesting ways for the design of more efficient fluorogenic G4 probes.
荧光检测DNA和RNA g -四重复合物(G4s)是一种非常有效的策略,不仅可以评估细胞G4s的存在和流行,还可以评估它们作为治疗干预目标的相关性。在用于此目的的荧光团中,开启探针是最有趣的,因为它们的荧光仅在与G4靶标相互作用时触发,这确保了检测的高灵敏度和选择性。我们报道了一系列双智能G4探针,它们既是智能G4配体(其结构在与G4s相互作用时重组),又是智能荧光探针(其荧光在与G4s相互作用时开启)。最佳原型N-TASQ优异性能背后的精细机械细节仍有待破译。为此,我们报道了两种类似物TzN-TASQ和AlkN-TASQ的合成和研究。对其G4相互作用特性的仔细分析,在体外和计算机上进行了研究,表明微调其组成结构元素可以提高其“关闭”(即低荧光构象)和“打开”状态(即高荧光构象)的效率,从而为设计更高效的荧光G4探针开辟了有趣的途径。
{"title":"Fluorescence Detection of DNA/RNA G-Quadruplexes (G4s) by Twice-as-Smart Ligands.","authors":"Jiri Ledvinka, Francesco Rota Sperti, Gabor Paragi, Marc Pirrotta, Nicolas Chéron, Ibai E Valverde, Petra Menova, David Monchaud","doi":"10.1002/cmdc.202400829","DOIUrl":"10.1002/cmdc.202400829","url":null,"abstract":"<p><p>Fluorescence detection of DNA and RNA G-quadruplexes (G4s) is a very efficient strategy to assess not only the existence and prevalence of cellular G4s but also their relevance as targets for therapeutic interventions. Among the fluorophores used to this end, turn-on probes are the most interesting since their fluorescence is triggered only upon interaction with their G4 targets, which ensures a high sensitivity and selectivity of detection. We reported on a series of twice-as-smart G4 probes, which are both smart G4 ligands (whose structure is reorganized upon interaction with G4s) and smart fluorescent probes (whose fluorescence is turned on upon interaction with G4s). The fine mechanistic details behind the excellent properties of the best prototype N-TASQ remain to be deciphered: to investigate this, we report here on the synthesis and studies of two analogues, <sup>Tz</sup>N-TASQ and <sup>Alk</sup>N-TASQ, and on a careful analysis of their G4-interacting properties, investigated both in vitro and in silico. Our results show that fine-tuning their constitutive structural elements allows for increasing the efficiency of both their 'off' (i. e., a conformation with a low fluorescence) and 'on' states (i. e., a conformation with a high fluorescence), which opens interesting ways for the design of more efficient fluorogenic G4 probes.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202400829"},"PeriodicalIF":3.6,"publicationDate":"2024-12-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142875569","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}
Deuterated drugs, which are derived from the subtle exchange of a protium atom with a deuterium atom in drug molecules, exhibit significant differences in pharmaceutical characteristics compared to their parent drugs. With the advantages of improving pharmacokinetic properties, reducing toxicity, inhibiting the interconversion between chiral drugs and restricting drug interactions, deuterated drugs have attracted widespread attention from medicinal chemists. This review highlights the application of deuteration strategies in drug design, summarizing the progress of all deuterated drugs available in the market or still under investigation to provide a reference for all researchers engaged deuterated drug development.
{"title":"The Application of Deuteration Strategy in Drug Design.","authors":"Yuzhu Chen, Yunfei Du","doi":"10.1002/cmdc.202400836","DOIUrl":"10.1002/cmdc.202400836","url":null,"abstract":"<p><p>Deuterated drugs, which are derived from the subtle exchange of a protium atom with a deuterium atom in drug molecules, exhibit significant differences in pharmaceutical characteristics compared to their parent drugs. With the advantages of improving pharmacokinetic properties, reducing toxicity, inhibiting the interconversion between chiral drugs and restricting drug interactions, deuterated drugs have attracted widespread attention from medicinal chemists. This review highlights the application of deuteration strategies in drug design, summarizing the progress of all deuterated drugs available in the market or still under investigation to provide a reference for all researchers engaged deuterated drug development.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202400836"},"PeriodicalIF":3.6,"publicationDate":"2024-12-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142880910","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}
Juraj Velcicky, Estelle Ngo, Matthias R Bauer, Arndt Meyer, Achim Schlapbach, Sophie Racine, David Orain, Daniel Pflieger, Sylvie Teixeira-Fouchard, Celine Dubois, Alban Goetz, Roland Steiner, Marco Palmieri, Alex Bussenault, Rowan Stringer, Patrice Larger, Simone Riek, Patrick Schmutz, Sylvie Lehmann, Clemens Scheufler, Jean-Michel Rondeau, Christoph Burkhart, Thomas Knoepfel, Nina Gommermann
The pro-inflammatory cytokine interleukin-17A (IL-17) plays an important role in the body's defense against bacterial and fungal infections. However, overexpression of IL-17 has been associated with several diseases, including rheumatoid arthritis, asthma, psoriasis, and even cancer. The role of IL-17 in psoriasis has been confirmed by clinical use of IL-17 antibodies, e. g. secukinumab (Cosentyx®). Ongoing research is focused on discovering low molecular weight IL-17 inhibitors. In this publication, we present thiazole-based IL-17 inhibitors discovered through a scaffold-morphing strategy. This strategy involved ring-opening of a known scaffold and utilization of a chalcogen interaction between thiazole-sulfur and central amide-oxygen to maintain the coplanar conformation found in the parent compound. The new scaffold enabled the generation of highly potent compounds with good overall profile. The optimized compounds 11 and 15 demonstrated good exposure in rats after oral dosing. Importantly, compound 11 exhibited no adverse effects in a rat tolerability study after a four-day administration of up to 300 mg/kg/day.
{"title":"Thiazole-Based IL-17 Inhibitors Discovered by Scaffold Morphing.","authors":"Juraj Velcicky, Estelle Ngo, Matthias R Bauer, Arndt Meyer, Achim Schlapbach, Sophie Racine, David Orain, Daniel Pflieger, Sylvie Teixeira-Fouchard, Celine Dubois, Alban Goetz, Roland Steiner, Marco Palmieri, Alex Bussenault, Rowan Stringer, Patrice Larger, Simone Riek, Patrick Schmutz, Sylvie Lehmann, Clemens Scheufler, Jean-Michel Rondeau, Christoph Burkhart, Thomas Knoepfel, Nina Gommermann","doi":"10.1002/cmdc.202400851","DOIUrl":"10.1002/cmdc.202400851","url":null,"abstract":"<p><p>The pro-inflammatory cytokine interleukin-17A (IL-17) plays an important role in the body's defense against bacterial and fungal infections. However, overexpression of IL-17 has been associated with several diseases, including rheumatoid arthritis, asthma, psoriasis, and even cancer. The role of IL-17 in psoriasis has been confirmed by clinical use of IL-17 antibodies, e. g. secukinumab (Cosentyx<sup>®</sup>). Ongoing research is focused on discovering low molecular weight IL-17 inhibitors. In this publication, we present thiazole-based IL-17 inhibitors discovered through a scaffold-morphing strategy. This strategy involved ring-opening of a known scaffold and utilization of a chalcogen interaction between thiazole-sulfur and central amide-oxygen to maintain the coplanar conformation found in the parent compound. The new scaffold enabled the generation of highly potent compounds with good overall profile. The optimized compounds 11 and 15 demonstrated good exposure in rats after oral dosing. Importantly, compound 11 exhibited no adverse effects in a rat tolerability study after a four-day administration of up to 300 mg/kg/day.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202400851"},"PeriodicalIF":3.6,"publicationDate":"2024-12-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142880925","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}
Butyrylcholinesterase plays an indispensable role in organisms, and its abnormal expression poses a significant threat to human health and safety, covering various aspects including liver-related diseases, diabetes, obesity, cardiovascular and cerebrovascular diseases, and neurodegenerative diseases. In addition, toxic substances such as organophosphorus and carbamate pesticides markedly inhibit BChE activity. BChE activity serves as a critical parameter for the clinical diagnosis of acute organophosphorus pesticide poisoning and the evaluation of organophosphorus and carbamate pesticide residues. Therefore, the accurate and reliable detection of butyrylcholinesterase activity is particularly urgent and important for in-depth analysis of its biological function, diagnosis and therapy of related diseases, drug screening and sensitive detection of pesticide residues. Fluorescent probes have become a promising tool for sensing and imaging of butyrylcholinesterase, due to its advantages of high spatio-temporal resolution, high selectivity, non-invasive, high sensitivity, and tailored molecule structures. Here, this paper provides a comprehensive overview of the research progress in the sensing, imaging and therapy of butyrylcholinesterase utilizing fluorescent probes. This paper might be a useful guideline for researchers to design new high-performance fluorescence probes for BChE, and making further contributions to this intriguing field.
{"title":"Small-Molecule Fluorescent Probes for Butyrylcholinesterase.","authors":"Zibo Lin, Yuanyuan Liao, Donglei Tian, Junyu Liao, Qiong Chen, Jun Yin","doi":"10.1002/cmdc.202400875","DOIUrl":"10.1002/cmdc.202400875","url":null,"abstract":"<p><p>Butyrylcholinesterase plays an indispensable role in organisms, and its abnormal expression poses a significant threat to human health and safety, covering various aspects including liver-related diseases, diabetes, obesity, cardiovascular and cerebrovascular diseases, and neurodegenerative diseases. In addition, toxic substances such as organophosphorus and carbamate pesticides markedly inhibit BChE activity. BChE activity serves as a critical parameter for the clinical diagnosis of acute organophosphorus pesticide poisoning and the evaluation of organophosphorus and carbamate pesticide residues. Therefore, the accurate and reliable detection of butyrylcholinesterase activity is particularly urgent and important for in-depth analysis of its biological function, diagnosis and therapy of related diseases, drug screening and sensitive detection of pesticide residues. Fluorescent probes have become a promising tool for sensing and imaging of butyrylcholinesterase, due to its advantages of high spatio-temporal resolution, high selectivity, non-invasive, high sensitivity, and tailored molecule structures. Here, this paper provides a comprehensive overview of the research progress in the sensing, imaging and therapy of butyrylcholinesterase utilizing fluorescent probes. This paper might be a useful guideline for researchers to design new high-performance fluorescence probes for BChE, and making further contributions to this intriguing field.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202400875"},"PeriodicalIF":3.6,"publicationDate":"2024-12-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142875525","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}
Aixa M Orta Rivera, Luis A Landrau Correa, Selene L Schiavone-Chamorro, Moriana Rankins, Mariela V Pérez Otero, Josué A Benjamín-Rivera, José A Vega Aponte, Valerie B Ebenki, Adriana I Vargas Figueroa, Andrei V Astashkin, Lauren Fernández-Vega, Arthur D Tinoco
Tinoco A-Team Deferasirox (Def), an orally administered iron-chelating drug, has drawn significant interest in repurposing for anticancer application due to the elevated Fe demand by cancer cells. But there are also concerns about its severe off target health effects. Herein Cu(II) binding is studied as a potential off target interaction. The aqueous solution stability and speciation of the ternary complex Cu(Def)(pyridine) was studied by UV-Vis and EPR spectroscopy, ESI-mass spectrometry, and cyclic voltammetry under physiologically relevant conditions. The complex is observed to be a redox active, mononuclear Cu(II) complex in square planar geometry. UV-Vis spectroscopy demonstrates that at pH 7.4 the complex is quite stable (ϵ337nm=10,820 M-1 cm-1) with a log K=16.65±0.1. Cu scavenging from the Cu transporters ceruloplasmin and albumin was also studied. Def does not inhibit ceruloplasmin activity but forms a ternary Cu(II) complex at the bovine serum albumin ATCUN site. Cu(Def)(py) displays potent but nonselective cytotoxicity against A549 cancer and MRC-5 noncancer lung cells but the potency of the ternary protein complex was more moderate. This work elucidates potential Def toxicity from Cu complexation in the body but also cytotoxic synergy between the metal and chelator that informs on new drug design directions.
由于癌细胞对铁的需求增加,一种口服铁螯合剂药物铁asirox (Def)已经引起了人们对抗癌应用的极大兴趣。但也有人担心其严重偏离目标的健康影响。本文将Cu(II)结合作为潜在的脱靶相互作用进行研究。在生理相关条件下,采用紫外可见光谱、EPR光谱、esi质谱和循环伏安法研究了三元配合物Cu(Def)(吡啶)的水溶液稳定性和形态。该配合物为具有氧化还原活性的单核Cu(II)配合物,呈方形平面几何结构。紫外可见光谱分析表明,在pH 7.4条件下,该配合物非常稳定(ε337nm = 10,820 M-1cm-1), log K = 16.65±0.1。铜转运体铜蓝蛋白和白蛋白对铜的清除也进行了研究。Def不抑制铜蓝蛋白活性,但在牛血清白蛋白ATCUN位点形成三元Cu(II)复合物。Cu(Def)(py)对A549癌和MRC-5非癌肺细胞表现出有效但非选择性的细胞毒性,但三元蛋白复合物的效力较中等。这项工作阐明了体内铜络合的潜在毒性,以及金属和螯合剂之间的细胞毒性协同作用,为新药设计方向提供了信息。
{"title":"Elucidating the High Affinity Copper(II) Complexation by the Iron Chelator Deferasirox Provides Therapeutic and Toxicity Insight.","authors":"Aixa M Orta Rivera, Luis A Landrau Correa, Selene L Schiavone-Chamorro, Moriana Rankins, Mariela V Pérez Otero, Josué A Benjamín-Rivera, José A Vega Aponte, Valerie B Ebenki, Adriana I Vargas Figueroa, Andrei V Astashkin, Lauren Fernández-Vega, Arthur D Tinoco","doi":"10.1002/cmdc.202400937","DOIUrl":"10.1002/cmdc.202400937","url":null,"abstract":"<p><p>Tinoco A-Team Deferasirox (Def), an orally administered iron-chelating drug, has drawn significant interest in repurposing for anticancer application due to the elevated Fe demand by cancer cells. But there are also concerns about its severe off target health effects. Herein Cu(II) binding is studied as a potential off target interaction. The aqueous solution stability and speciation of the ternary complex Cu(Def)(pyridine) was studied by UV-Vis and EPR spectroscopy, ESI-mass spectrometry, and cyclic voltammetry under physiologically relevant conditions. The complex is observed to be a redox active, mononuclear Cu(II) complex in square planar geometry. UV-Vis spectroscopy demonstrates that at pH 7.4 the complex is quite stable (ϵ<sub>337nm</sub>=10,820 M<sup>-1</sup> cm<sup>-1</sup>) with a log K=16.65±0.1. Cu scavenging from the Cu transporters ceruloplasmin and albumin was also studied. Def does not inhibit ceruloplasmin activity but forms a ternary Cu(II) complex at the bovine serum albumin ATCUN site. Cu(Def)(py) displays potent but nonselective cytotoxicity against A549 cancer and MRC-5 noncancer lung cells but the potency of the ternary protein complex was more moderate. This work elucidates potential Def toxicity from Cu complexation in the body but also cytotoxic synergy between the metal and chelator that informs on new drug design directions.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202400937"},"PeriodicalIF":3.6,"publicationDate":"2024-12-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142880907","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}
Jaka Kranjc, Tihomir Tomašič, Stane Pajk, Matjaž Brinc, Anja Pišlar, Marko Anderluh
Seven different enzymes comprise the galactosyltransferases family, of which β-1,4-galactosyltransferase I (β-1,4-GALT1) is the major contributor to galactosylation activity in cells. Since abnormalities in galactosylation are associated with many pathophysiological conditions, β-1,4-GALT1 is an interesting new target for drug discovery and molecular probe design. There are several known β-1,4-GALT1 inhibitors, but most of them suffer from low cell permeability and thus low in vivo activity. In the present work, we describe an in silico screening performed using commercially available virtual compound libraries that led us to the discovery of novel β-1,4-GALT1 inhibitors. A virtual screening campaign was performed by docking compound libraries to the binding site of β-1,4-GALT1, followed by biological evaluation of selected hits for their β-1,4-GALT1 inhibitory activity. The IC50 values were determined for the best performing inhibitors to obtain new chemotypes of β-1,4-GALT1 inhibitors.
{"title":"New Inhibitors of β-1,4-Galactosyltransferase I Discovered by Virtual Screening.","authors":"Jaka Kranjc, Tihomir Tomašič, Stane Pajk, Matjaž Brinc, Anja Pišlar, Marko Anderluh","doi":"10.1002/cmdc.202400896","DOIUrl":"10.1002/cmdc.202400896","url":null,"abstract":"<p><p>Seven different enzymes comprise the galactosyltransferases family, of which β-1,4-galactosyltransferase I (β-1,4-GALT1) is the major contributor to galactosylation activity in cells. Since abnormalities in galactosylation are associated with many pathophysiological conditions, β-1,4-GALT1 is an interesting new target for drug discovery and molecular probe design. There are several known β-1,4-GALT1 inhibitors, but most of them suffer from low cell permeability and thus low in vivo activity. In the present work, we describe an in silico screening performed using commercially available virtual compound libraries that led us to the discovery of novel β-1,4-GALT1 inhibitors. A virtual screening campaign was performed by docking compound libraries to the binding site of β-1,4-GALT1, followed by biological evaluation of selected hits for their β-1,4-GALT1 inhibitory activity. The IC<sub>50</sub> values were determined for the best performing inhibitors to obtain new chemotypes of β-1,4-GALT1 inhibitors.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202400896"},"PeriodicalIF":3.6,"publicationDate":"2024-12-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142875592","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}
Yiqing Fan, Zeqi Zeng, Jiaxian Mo, Zike Wang, Hongyu Jiang, Juanjuan Liu, Hai Qian, Wei Shi
The activation of the STING-mediated signaling pathway leads to the secretion of type I interferon (IFN) and the activation of tumor-specific T cells. STING, a pattern recognition receptor located on the endoplasmic reticulum membrane of immune cells, binds with endogenous cyclic dinucleotides. STING undergoes phosphorylation, triggering the STING-TBK1-IRF3 pathway and NF-κB pathway, resulting in the release of IFN-β and other pro-inflammatory cytokines, ultimately enhancing the activation of tumor-specific T cells. This mechanism serves to complement the limitations of immune checkpoint inhibitors and enhances the efficiency of the immune response. This study selected benzimidazole compounds GSK and SR-717, which exhibit promising potential as patented medicines, as our lead compounds. Aiming to address the challenges associated with the short half-life of benzimidazole compounds and the limited molecular activity of SR-717, we designed and synthesized a series of STING agonists (compounds 6~29). The compound 17 showed excellent agonistic activity on hSTING protein in vitro. The cytotoxicity tests of all the synthesized compounds were performed in vitro. Performed in vivo pharmacokinetic studies on the most promising compounds and conducted molecular docking analyses.
{"title":"Design, Synthesis, and Biological Assessment of Novel Aminobenzidazole Agonists Targeting the Stimulator of Interferon Genes (STING) Receptor Signaling Pathway for Oncology Immunotherapy.","authors":"Yiqing Fan, Zeqi Zeng, Jiaxian Mo, Zike Wang, Hongyu Jiang, Juanjuan Liu, Hai Qian, Wei Shi","doi":"10.1002/cmdc.202400695","DOIUrl":"10.1002/cmdc.202400695","url":null,"abstract":"<p><p>The activation of the STING-mediated signaling pathway leads to the secretion of type I interferon (IFN) and the activation of tumor-specific T cells. STING, a pattern recognition receptor located on the endoplasmic reticulum membrane of immune cells, binds with endogenous cyclic dinucleotides. STING undergoes phosphorylation, triggering the STING-TBK1-IRF3 pathway and NF-κB pathway, resulting in the release of IFN-β and other pro-inflammatory cytokines, ultimately enhancing the activation of tumor-specific T cells. This mechanism serves to complement the limitations of immune checkpoint inhibitors and enhances the efficiency of the immune response. This study selected benzimidazole compounds GSK and SR-717, which exhibit promising potential as patented medicines, as our lead compounds. Aiming to address the challenges associated with the short half-life of benzimidazole compounds and the limited molecular activity of SR-717, we designed and synthesized a series of STING agonists (compounds 6~29). The compound 17 showed excellent agonistic activity on hSTING protein in vitro. The cytotoxicity tests of all the synthesized compounds were performed in vitro. Performed in vivo pharmacokinetic studies on the most promising compounds and conducted molecular docking analyses.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202400695"},"PeriodicalIF":3.6,"publicationDate":"2024-12-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142875557","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}
Lhana Tisseur, Sandrine Cojean, Khadidiatou Gassama, Cédric Logé, Fabrice Pagniez, Christian Cavé, Guillaume Bernadat, Philippe M Loiseau, Stéphane Bach, Jérôme Thiéfaine, Carine Picot, Christophe Tomasoni, Olivier Leclercq, Blandine Baratte, Thomas Robert, Patrice Le Pape, Najma Rachidi, Marc-Antoine Bazin, Pascal Marchand
Our research group previously discovered CTN1122, an imidazo[1,2-a]pyrazine compound with promising antileishmanial activity against intramacrophage amastigotes of Leishmania major and L. donovani strains. CTN1122 effectively targets Leishmania casein kinase 1 (L-CK1.2) and exhibits a favorable safety profile. To further explore its chemical space, we developed a convergent strategy to modify the C2 position of the imidazo[1,2-a]pyrazine core using Suzuki-Miyaura coupling of the corresponding triflate intermediate. Among 15 newly synthesized analogs, seven derivatives featuring variously substituted phenyl rings at C2 demonstrated L-CK1.2 inhibition within micromolar to submicromolar ranges and antileishmanial activity in vitro with low cytotoxicity in macrophages. Compounds 7 d and 7 l were particularly potent, with IC50 values of 1.25 μM and 0.92 μM against L. major, and 1.44 μM and 2.34 μM against L. donovani, respectively. They showed IC50 L-CK1.2=0.30 μM and 0.57 μM with enhanced selectivity indices (SI=3.8 and 1.6) over the human CK1ϵ ortholog. Additionally, four C2 analogs and two C5 isomers exhibited notable antiparasitic effects without strongly inhibiting L-CK1.2, indicating a possible alternative mechanism of action. Compound 7 k displayed the highest general activity, with IC50 values of 0.31 μM on L. major and 0.27 μM on L. donovani, coupled with favorable selectivity indexes.
{"title":"Investigating the C2 Modulation of the Imidazo[1,2-a]pyrazine-Based Hit Compound CTN1122: Synthesis, in vitro Antileishmanial Activity, Cytotoxicity and Casein Kinase 1 Inhibition.","authors":"Lhana Tisseur, Sandrine Cojean, Khadidiatou Gassama, Cédric Logé, Fabrice Pagniez, Christian Cavé, Guillaume Bernadat, Philippe M Loiseau, Stéphane Bach, Jérôme Thiéfaine, Carine Picot, Christophe Tomasoni, Olivier Leclercq, Blandine Baratte, Thomas Robert, Patrice Le Pape, Najma Rachidi, Marc-Antoine Bazin, Pascal Marchand","doi":"10.1002/cmdc.202400862","DOIUrl":"10.1002/cmdc.202400862","url":null,"abstract":"<p><p>Our research group previously discovered CTN1122, an imidazo[1,2-a]pyrazine compound with promising antileishmanial activity against intramacrophage amastigotes of Leishmania major and L. donovani strains. CTN1122 effectively targets Leishmania casein kinase 1 (L-CK1.2) and exhibits a favorable safety profile. To further explore its chemical space, we developed a convergent strategy to modify the C2 position of the imidazo[1,2-a]pyrazine core using Suzuki-Miyaura coupling of the corresponding triflate intermediate. Among 15 newly synthesized analogs, seven derivatives featuring variously substituted phenyl rings at C2 demonstrated L-CK1.2 inhibition within micromolar to submicromolar ranges and antileishmanial activity in vitro with low cytotoxicity in macrophages. Compounds 7 d and 7 l were particularly potent, with IC<sub>50</sub> values of 1.25 μM and 0.92 μM against L. major, and 1.44 μM and 2.34 μM against L. donovani, respectively. They showed IC<sub>50</sub> L-CK1.2=0.30 μM and 0.57 μM with enhanced selectivity indices (SI=3.8 and 1.6) over the human CK1ϵ ortholog. Additionally, four C2 analogs and two C5 isomers exhibited notable antiparasitic effects without strongly inhibiting L-CK1.2, indicating a possible alternative mechanism of action. Compound 7 k displayed the highest general activity, with IC<sub>50</sub> values of 0.31 μM on L. major and 0.27 μM on L. donovani, coupled with favorable selectivity indexes.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202400862"},"PeriodicalIF":3.6,"publicationDate":"2024-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142833264","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}
Shravani Battula, Haripriya Bhumannagari, S S S S Sudha Ambadipudi, Sai Balaji Andugulapati, Kiranmai Nayani
An efficient and concise synthesis of highly functionalized bridged coumarins has been developed through a diastereoselective double Michael addition reaction of p-quinols with various 4-hydroxy coumarins under catalyst-free conditions in H2O-DMSO (8 : 2). The method has been applied to oxindoles for the synthesis of a variety of bridged-oxindoles and bridged-spiroxindoles in presence of a DABCO base using H2O-EtOH (8 : 2) as solvent medium. The strategy is simple, highly atom economical as there is no by-product and environmentally benign (E-factor=0.1-0.9). The synthesized compounds were screened against triple-negative breast cancers and found that bridged coumarin (3 a) and oxindole (5 d) compounds exhibit potent anti-cancer activity at 6.6 and 8.8 μM (IC50) concentrations respectively. Further analysis revealed that 3 a and 5 d caused elevated early and total apoptosis by arresting the MDA-MB-468 cells in G2/M phase of the cell cycle. Overall, our results demonstrate that bridged coumarin (3 a) and oxindole (5 d) compounds-based approach attenuates the cancer progression and may pave a path for the translational outcome.
{"title":"Diastereoselective Cascade Double Michael Addition to Access Bridged Coumarins, Oxindoles and Spirooxindoles: A Sustainable Strategy for Synthesis of Anticancer Molecules.","authors":"Shravani Battula, Haripriya Bhumannagari, S S S S Sudha Ambadipudi, Sai Balaji Andugulapati, Kiranmai Nayani","doi":"10.1002/cmdc.202400946","DOIUrl":"10.1002/cmdc.202400946","url":null,"abstract":"<p><p>An efficient and concise synthesis of highly functionalized bridged coumarins has been developed through a diastereoselective double Michael addition reaction of p-quinols with various 4-hydroxy coumarins under catalyst-free conditions in H<sub>2</sub>O-DMSO (8 : 2). The method has been applied to oxindoles for the synthesis of a variety of bridged-oxindoles and bridged-spiroxindoles in presence of a DABCO base using H<sub>2</sub>O-EtOH (8 : 2) as solvent medium. The strategy is simple, highly atom economical as there is no by-product and environmentally benign (E-factor=0.1-0.9). The synthesized compounds were screened against triple-negative breast cancers and found that bridged coumarin (3 a) and oxindole (5 d) compounds exhibit potent anti-cancer activity at 6.6 and 8.8 μM (IC<sub>50</sub>) concentrations respectively. Further analysis revealed that 3 a and 5 d caused elevated early and total apoptosis by arresting the MDA-MB-468 cells in G2/M phase of the cell cycle. Overall, our results demonstrate that bridged coumarin (3 a) and oxindole (5 d) compounds-based approach attenuates the cancer progression and may pave a path for the translational outcome.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202400946"},"PeriodicalIF":3.6,"publicationDate":"2024-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142833262","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}
Baptiste Dupouy, Tanja Karpstein, Cécile Häberli, Monica Cal, Matthias Rottmann, Pascal Mäser, Jennifer Keiser, B Cichocki, Mourad Elhabiri, Elisabeth Davioud-Charvet
This study explores the synthesis and evaluation of novel 1,2,3-triazole-methyl-1,4-naphthoquinone hybrids, focusing on their electrochemical properties and antiparasitic efficacies against two human blood-dwelling parasites Plasmodium falciparum and Schistosoma mansoni. Using copper-catalyzed azide-alkyne cycloaddition (CuAAC), a well-established tool in click chemistry, two synthetic routes were assessed to develop α- and β-[triazole-methyl]-menadione derivatives. By optimizing the CuAAC reaction conditions, yields were significantly improved, reaching up to 94 % for key intermediates and resulting in the formation of a library of approximately 30 compounds. Biological evaluation of the compounds in antiparasitic drug assays demonstrated notable antischistosomal potencies, while no significant activity was observed for the same series against P. falciparum parasites. Electrochemical and 'benzylic' oxidation studies confirmed that the active 'benzoyl' metabolite responsible for the antiplasmodial activity of plasmodione cannot be generated. These findings highlight the potential of triazole-linked menadione hybrids as promising early candidates for antischistosomal drug development, and provides insights into structure-activity relationships crucial for future therapeutic strategies.
{"title":"Synthesis of 1,2,3-Triazole-Methyl-Menadione Derivatives: Evaluation of Electrochemical and Antiparasitic Properties against two Blood-Dwelling Parasites.","authors":"Baptiste Dupouy, Tanja Karpstein, Cécile Häberli, Monica Cal, Matthias Rottmann, Pascal Mäser, Jennifer Keiser, B Cichocki, Mourad Elhabiri, Elisabeth Davioud-Charvet","doi":"10.1002/cmdc.202400731","DOIUrl":"10.1002/cmdc.202400731","url":null,"abstract":"<p><p>This study explores the synthesis and evaluation of novel 1,2,3-triazole-methyl-1,4-naphthoquinone hybrids, focusing on their electrochemical properties and antiparasitic efficacies against two human blood-dwelling parasites Plasmodium falciparum and Schistosoma mansoni. Using copper-catalyzed azide-alkyne cycloaddition (CuAAC), a well-established tool in click chemistry, two synthetic routes were assessed to develop α- and β-[triazole-methyl]-menadione derivatives. By optimizing the CuAAC reaction conditions, yields were significantly improved, reaching up to 94 % for key intermediates and resulting in the formation of a library of approximately 30 compounds. Biological evaluation of the compounds in antiparasitic drug assays demonstrated notable antischistosomal potencies, while no significant activity was observed for the same series against P. falciparum parasites. Electrochemical and 'benzylic' oxidation studies confirmed that the active 'benzoyl' metabolite responsible for the antiplasmodial activity of plasmodione cannot be generated. These findings highlight the potential of triazole-linked menadione hybrids as promising early candidates for antischistosomal drug development, and provides insights into structure-activity relationships crucial for future therapeutic strategies.</p>","PeriodicalId":147,"journal":{"name":"ChemMedChem","volume":" ","pages":"e202400731"},"PeriodicalIF":3.6,"publicationDate":"2024-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142826827","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}