A series of sulfonamides incorporating a 1,2,3-triazolyloxime substituted 1,2,3-triazolyl moiety were conceptualized and synthesized as human carbonic anhydrase (hCA) inhibitors. The synthesized small structures, denoted 7a through 7o, exhibited moderate inhibitory effects against the tumor-associated isoforms hCA IX and hCA XII compared to the well-known hCA inhibitor acetazolamide. In contrast, these molecules demonstrated higher potency and a diverse range of selectivity against the cytosolic isoforms hCA I and hCA II. Notably, the 4-hydroxyphenyl derivative (compound 7dversus cytosolic isoforms), the 4-acetylphenyl derivative (compound 7o), and the phenyl derivative (compound 7a) emerged as the most potent and selective inhibitors in this series, with inhibition constants (KI) of 47.1, 35.9, 170.0, and 149.9 nM, respectively, against hCA I, II, IX, and XII. Further cytotoxicity assays of compounds 7a-o against cancer cell lines Hep3B and A549, as well as normal cell line L929, were conducted to assess their selectivity towards malignant cells. Compounds 7d, 7g, and 7k exhibited selective cytotoxicity towards the Hep3B cell line, with reduced selectivity towards A549, whereas compound 7j demonstrated higher selectivity for the A549 cell line. Additionally, molecular docking studies were performed to elucidate the binding modes of these compounds within the active sites of hCAs, revealing crucial interactions that underpin their significant activity and selectivity for the tumor-specific isoforms.
{"title":"Novel benzenesulfonamides containing a dual triazole moiety with selective carbonic anhydrase inhibition and anticancer activity.","authors":"Aida Buza, Cüneyt Türkeş, Mustafa Arslan, Yeliz Demir, Busra Dincer, Arleta Rifati Nixha, Şükrü Beydemir","doi":"10.1039/d4md00617h","DOIUrl":"10.1039/d4md00617h","url":null,"abstract":"<p><p>A series of sulfonamides incorporating a 1,2,3-triazolyloxime substituted 1,2,3-triazolyl moiety were conceptualized and synthesized as human carbonic anhydrase (<i>h</i>CA) inhibitors. The synthesized small structures, denoted 7a through 7o, exhibited moderate inhibitory effects against the tumor-associated isoforms <i>h</i>CA IX and <i>h</i>CA XII compared to the well-known <i>h</i>CA inhibitor acetazolamide. In contrast, these molecules demonstrated higher potency and a diverse range of selectivity against the cytosolic isoforms <i>h</i>CA I and <i>h</i>CA II. Notably, the 4-hydroxyphenyl derivative (compound 7d<i>versus</i> cytosolic isoforms), the 4-acetylphenyl derivative (compound 7o), and the phenyl derivative (compound 7a) emerged as the most potent and selective inhibitors in this series, with inhibition constants (<i>K</i> <sub>I</sub>) of 47.1, 35.9, 170.0, and 149.9 nM, respectively, against <i>h</i>CA I, II, IX, and XII. Further cytotoxicity assays of compounds 7a-o against cancer cell lines Hep3B and A549, as well as normal cell line L929, were conducted to assess their selectivity towards malignant cells. Compounds 7d, 7g, and 7k exhibited selective cytotoxicity towards the Hep3B cell line, with reduced selectivity towards A549, whereas compound 7j demonstrated higher selectivity for the A549 cell line. Additionally, molecular docking studies were performed to elucidate the binding modes of these compounds within the active sites of <i>h</i>CAs, revealing crucial interactions that underpin their significant activity and selectivity for the tumor-specific isoforms.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2024-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11525713/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142568514","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}
Arianna Amenta, Susanna Comi, Marcelo Kravicz, Silvia Sesana, Antonia Antoniou, Daniele Passarella, Pierfausto Seneci, Sara Pellegrino, Francesca Re
Pimasertib, a potent antiproliferative drug, has been extensively studied for treating cancers characterized by dysregulation in the ERK/MAPK signaling pathway, such as melanoma. However, its therapeutic efficacy would greatly benefit from an increased selectivity for tumour cells and a longer half-life. Such improvements may be achieved by combining the rational design of a prodrug with its encapsulation in a potential nanodelivery system. For this reason, we synthesized a glutathione (GSH)-responsive putative prodrug of pimasertib (PROPIMA), which contains a redox-sensitive disulphide linker that can be processed by GSH to activate pimasertib. The synthesis of PROPIMA and its in vitro biological activity on a human melanoma cell line as a model are described. The results showed that PROPIMA, either free or embedded in liposomes, selectively inhibits cell proliferation and cell viability, reducing by about 5-fold the levels of pERK. Additionally, PROPIMA shows stronger inhibition of the cancer cell migration than the parent drug.
{"title":"A novel, glutathione-activated prodrug of pimasertib loaded in liposomes for targeted cancer therapy.","authors":"Arianna Amenta, Susanna Comi, Marcelo Kravicz, Silvia Sesana, Antonia Antoniou, Daniele Passarella, Pierfausto Seneci, Sara Pellegrino, Francesca Re","doi":"10.1039/d4md00517a","DOIUrl":"10.1039/d4md00517a","url":null,"abstract":"<p><p>Pimasertib, a potent antiproliferative drug, has been extensively studied for treating cancers characterized by dysregulation in the ERK/MAPK signaling pathway, such as melanoma. However, its therapeutic efficacy would greatly benefit from an increased selectivity for tumour cells and a longer half-life. Such improvements may be achieved by combining the rational design of a prodrug with its encapsulation in a potential nanodelivery system. For this reason, we synthesized a glutathione (GSH)-responsive putative prodrug of pimasertib (PROPIMA), which contains a redox-sensitive disulphide linker that can be processed by GSH to activate pimasertib. The synthesis of PROPIMA and its <i>in vitro</i> biological activity on a human melanoma cell line as a model are described. The results showed that PROPIMA, either free or embedded in liposomes, selectively inhibits cell proliferation and cell viability, reducing by about 5-fold the levels of pERK. Additionally, PROPIMA shows stronger inhibition of the cancer cell migration than the parent drug.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2024-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11485093/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142473751","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}
Alzheimer's disease (AD) is a prevalent degenerative disorder affecting the central nervous system of the elderly. Patients primarily manifest cognitive decline and non-cognitive neuro-psychiatric symptoms. Currently, western medications for AD primarily include cholinesterase inhibitors and glutamate receptor inhibitors, which have limited efficacy and accompanied by significant toxic side effects. Given the intricate pathogenesis of AD, the use of single-target inhibitors is limited. In recent years, as research on AD has progressed, traditional Chinese medicine (TCM) and its active ingredients have increasingly played a crucial role in clinical treatment. Numerous studies demonstrate that TCM and its active ingredients can exert anti-Alzheimer's effects by modulating pathological protein production and deposition, inhibiting tau protein hyperphosphorylation, apoptosis, inflammation, and oxidative stress, while enhancing the central cholinergic system, protecting neurons and synapses, and optimizing energy metabolism. This article summarizes extracts from TCM and briefly elucidates their pharmacological mechanisms against AD, aiming to provide a foundation for further research into the specific mechanisms of TCM in the prevention and treatment of the disease, as well as the identification of efficacious active ingredients.
阿尔茨海默病(AD)是一种影响老年人中枢神经系统的常见退行性疾病。患者主要表现为认知能力下降和非认知性神经精神症状。目前,治疗 AD 的西药主要包括胆碱酯酶抑制剂和谷氨酸受体抑制剂,但疗效有限,且毒副作用大。鉴于 AD 的发病机制错综复杂,单一靶点抑制剂的使用受到限制。近年来,随着对多发性硬化症研究的深入,中药及其有效成分在临床治疗中发挥着越来越重要的作用。大量研究表明,中药及其有效成分可通过调节病理蛋白的产生和沉积,抑制tau蛋白高磷酸化、细胞凋亡、炎症和氧化应激,同时增强中枢胆碱能系统,保护神经元和突触,优化能量代谢,从而发挥抗阿尔茨海默病的作用。本文对中药提取物进行了总结,并简要阐明了其防治AD的药理机制,旨在为进一步研究中药防治AD的具体机理以及确定有效的活性成分奠定基础。
{"title":"Pathogenesis of Alzheimer's disease and therapeutic strategies involving traditional Chinese medicine.","authors":"Shutang Li, Jinfei Yang","doi":"10.1039/d4md00660g","DOIUrl":"10.1039/d4md00660g","url":null,"abstract":"<p><p>Alzheimer's disease (AD) is a prevalent degenerative disorder affecting the central nervous system of the elderly. Patients primarily manifest cognitive decline and non-cognitive neuro-psychiatric symptoms. Currently, western medications for AD primarily include cholinesterase inhibitors and glutamate receptor inhibitors, which have limited efficacy and accompanied by significant toxic side effects. Given the intricate pathogenesis of AD, the use of single-target inhibitors is limited. In recent years, as research on AD has progressed, traditional Chinese medicine (TCM) and its active ingredients have increasingly played a crucial role in clinical treatment. Numerous studies demonstrate that TCM and its active ingredients can exert anti-Alzheimer's effects by modulating pathological protein production and deposition, inhibiting tau protein hyperphosphorylation, apoptosis, inflammation, and oxidative stress, while enhancing the central cholinergic system, protecting neurons and synapses, and optimizing energy metabolism. This article summarizes extracts from TCM and briefly elucidates their pharmacological mechanisms against AD, aiming to provide a foundation for further research into the specific mechanisms of TCM in the prevention and treatment of the disease, as well as the identification of efficacious active ingredients.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2024-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11484936/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142473776","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}
Stefano Tomassi, Benito Natale, Michele Roggia, Luisa Amato, Caterina De Rosa, Carminia Maria Della Corte, Emma Baglini, Giorgio Amendola, Anna Messere, Salvatore Di Maro, Elisabetta Barresi, Federico Da Settimo, Maria Letizia Trincavelli, Fortunato Ciardiello, Sabrina Taliani, Floriana Morgillo, Sandro Cosconati
Non-small cell lung cancer (NSCLC), the leading cause of cancer-related mortality worldwide, poses a formidable challenge due to its heterogeneity and the emergence of resistance to targeted therapies. While initially effective, first- and third-generation EGFR-tyrosine kinase inhibitors (TKIs) often fail to control disease progression, leaving patients with limited treatment options. To address this unmet medical need, we explored the therapeutic potential of multitargeting agents that simultaneously inhibit two key signalling pathways, the mesenchymal-epithelial transition factor (c-MET) and the G protein-coupled receptor Smoothened (SMO), frequently dysregulated in NSCLC. By employing a combination of in silico drug repurposing and structure-based structure-activity relationship (SAR) studies, we identified and developed novel c-MET/SMO-targeting agents with antiproliferative activity against first- as well as third-generation EGFR-TKI-resistant NSCLC cells suggesting a synergistic effect arising from the simultaneous inhibition of c-MET and SMO.
非小细胞肺癌(NSCLC)是全球癌症相关死亡的主要原因,由于其异质性和靶向疗法耐药性的出现,它构成了一项艰巨的挑战。第一代和第三代表皮生长因子受体酪氨酸激酶抑制剂(TKIs)虽然最初有效,但往往无法控制疾病进展,给患者留下的治疗选择非常有限。为了满足这一尚未满足的医疗需求,我们探索了多靶点药物的治疗潜力,这些药物可同时抑制两种关键信号通路,即间充质-上皮转化因子(c-MET)和G蛋白偶联受体SMO(Smoothened),这两种信号通路在NSCLC中经常失调。通过结合使用硅学药物再利用和基于结构的结构-活性关系(SAR)研究,我们发现并开发了新型 c-MET/SMO 靶向药物,它们对第一代和第三代表皮生长因子受体-TKI 抗性 NSCLC 细胞具有抗增殖活性,表明同时抑制 c-MET 和 SMO 可产生协同效应。
{"title":"Discovery of <i>N</i>-substituted-2-oxoindolin benzoylhydrazines as c-MET/SMO modulators in EGFRi-resistant non-small cell lung cancer.","authors":"Stefano Tomassi, Benito Natale, Michele Roggia, Luisa Amato, Caterina De Rosa, Carminia Maria Della Corte, Emma Baglini, Giorgio Amendola, Anna Messere, Salvatore Di Maro, Elisabetta Barresi, Federico Da Settimo, Maria Letizia Trincavelli, Fortunato Ciardiello, Sabrina Taliani, Floriana Morgillo, Sandro Cosconati","doi":"10.1039/d4md00553h","DOIUrl":"https://doi.org/10.1039/d4md00553h","url":null,"abstract":"<p><p>Non-small cell lung cancer (NSCLC), the leading cause of cancer-related mortality worldwide, poses a formidable challenge due to its heterogeneity and the emergence of resistance to targeted therapies. While initially effective, first- and third-generation EGFR-tyrosine kinase inhibitors (TKIs) often fail to control disease progression, leaving patients with limited treatment options. To address this unmet medical need, we explored the therapeutic potential of multitargeting agents that simultaneously inhibit two key signalling pathways, the mesenchymal-epithelial transition factor (c-MET) and the G protein-coupled receptor Smoothened (SMO), frequently dysregulated in NSCLC. By employing a combination of <i>in silico</i> drug repurposing and structure-based structure-activity relationship (SAR) studies, we identified and developed novel c-MET/SMO-targeting agents with antiproliferative activity against first- as well as third-generation EGFR-TKI-resistant NSCLC cells suggesting a synergistic effect arising from the simultaneous inhibition of c-MET and SMO.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2024-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11539002/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142606193","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}
Enveloped viruses enter the host cells by endocytosis and subsequently fuse with the endosomal membranes, or fuse with the plasma membrane at the cell surface. The crucial stage of viral infection, regardless of the route taken to enter the host cell, is membrane fusion. The present work aims to develop a peptide-based fusion inhibitor that prevents membrane fusion by modifying the properties of the participating membranes, without targeting a protein. This would allow us to develop a fusion inhibitor that might work against a larger spectrum of enveloped viruses as it does not target any specific viral fusion protein. With this goal in mind, we have designed a novel peptide by modifying a native sequence derived from coronin 1, a phagosomal protein, that helps to avoid lysosomal degradation of mycobacterium-loaded phagosomes. The designed peptide, mTG-23, inhibits ∼30-40% fusion between small unilamellar vesicles containing varying amounts of cholesterol by modulating the biophysical properties of the participating bilayers. As a proof of principle, we have further demonstrated that the mTG-23 inhibits Influenza A virus infection in A549 and MDCK cells (with ∼EC50 of 20.45 μM and 21.55 μM, respectively), where viral envelope and endosomal membrane fusion is a crucial step. Through a gamut of biophysical and biochemical methods, we surmise that mTG-23 inhibits viral infection by inhibiting viral envelope and endosomal membrane fusion. We envisage that the proposed antiviral strategy can be extended to other viruses that employ a similar modus operandi, providing a novel pan-antiviral approach.
{"title":"Developing peptide-based fusion inhibitors as an antiviral strategy utilizing coronin 1 as a template.","authors":"Manbit Subhadarsi Panda, Bushra Qazi, Vaishali Vishwakarma, Gourab Prasad Pattnaik, Sourav Haldar, Hirak Chakraborty","doi":"10.1039/d4md00523f","DOIUrl":"https://doi.org/10.1039/d4md00523f","url":null,"abstract":"<p><p>Enveloped viruses enter the host cells by endocytosis and subsequently fuse with the endosomal membranes, or fuse with the plasma membrane at the cell surface. The crucial stage of viral infection, regardless of the route taken to enter the host cell, is membrane fusion. The present work aims to develop a peptide-based fusion inhibitor that prevents membrane fusion by modifying the properties of the participating membranes, without targeting a protein. This would allow us to develop a fusion inhibitor that might work against a larger spectrum of enveloped viruses as it does not target any specific viral fusion protein. With this goal in mind, we have designed a novel peptide by modifying a native sequence derived from coronin 1, a phagosomal protein, that helps to avoid lysosomal degradation of mycobacterium-loaded phagosomes. The designed peptide, mTG-23, inhibits ∼30-40% fusion between small unilamellar vesicles containing varying amounts of cholesterol by modulating the biophysical properties of the participating bilayers. As a proof of principle, we have further demonstrated that the mTG-23 inhibits Influenza A virus infection in A549 and MDCK cells (with ∼EC<sub>50</sub> of 20.45 μM and 21.55 μM, respectively), where viral envelope and endosomal membrane fusion is a crucial step. Through a gamut of biophysical and biochemical methods, we surmise that mTG-23 inhibits viral infection by inhibiting viral envelope and endosomal membrane fusion. We envisage that the proposed antiviral strategy can be extended to other viruses that employ a similar modus operandi, providing a novel pan-antiviral approach.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11467784/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142473769","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}
David A Davis, Ashwin Nair, Yana Astter, Emma Treco, Brian Peyser, Rick Gussio, Tam Nguyen, Brett Eaton, Elena Postnikova, Michael Murphy, Prabha Shrestha, Haydar Bulut, Shin-Ichiro Hattorri, Hiroaki Mitsuya, Robert Yarchoan
Coronaviruses rely on the viral-encoded chymotrypsin-like main protease (Mpro or 3CLpro) for replication and assembly. Our previous research on Mpro of SARS-CoV-2 identified cysteine 300 (Cys300) as a potential allosteric site of Mpro inhibition. Here, we identified tixocortol (TX) as a covalent modifier of Cys300 which inhibits Mpro activity in vitro as well as in a cell-based Mpro expression assay. Most importantly TX inhibited SARS-CoV-2 replication in ACE2 expressing HeLa cells. Biochemical analysis and kinetic assays were consistent with TX acting as a non-competitive inhibitor. By contrast, TX was a weaker inhibitor and modifier of C300S Mpro, confirming a role for Cys300 in inhibition of WT Mpro but also providing evidence for an additional Cys target. TX pivalate (TP), a prodrug for TX that was previously marketed as a nasal spray, also inhibited SARS-CoV-2 replication in HeLa-ACE2 cells at low micromolar IC50s. These studies suggest that TX and/or TP could possibly be repurposed for the prevention and/or treatment of SARS-CoV-2 infection.
{"title":"Discovery of a nasal spray steroid, tixocortol, as an inhibitor of SARS-CoV-2 main protease and viral replication.","authors":"David A Davis, Ashwin Nair, Yana Astter, Emma Treco, Brian Peyser, Rick Gussio, Tam Nguyen, Brett Eaton, Elena Postnikova, Michael Murphy, Prabha Shrestha, Haydar Bulut, Shin-Ichiro Hattorri, Hiroaki Mitsuya, Robert Yarchoan","doi":"10.1039/d4md00454j","DOIUrl":"10.1039/d4md00454j","url":null,"abstract":"<p><p>Coronaviruses rely on the viral-encoded chymotrypsin-like main protease (M<sup>pro</sup> or 3CL<sup>pro</sup>) for replication and assembly. Our previous research on M<sup>pro</sup> of SARS-CoV-2 identified cysteine 300 (Cys300) as a potential allosteric site of M<sup>pro</sup> inhibition. Here, we identified tixocortol (TX) as a covalent modifier of Cys300 which inhibits M<sup>pro</sup> activity <i>in vitro</i> as well as in a cell-based M<sup>pro</sup> expression assay. Most importantly TX inhibited SARS-CoV-2 replication in ACE2 expressing HeLa cells. Biochemical analysis and kinetic assays were consistent with TX acting as a non-competitive inhibitor. By contrast, TX was a weaker inhibitor and modifier of C300S M<sup>pro</sup>, confirming a role for Cys300 in inhibition of WT M<sup>pro</sup> but also providing evidence for an additional Cys target. TX pivalate (TP), a prodrug for TX that was previously marketed as a nasal spray, also inhibited SARS-CoV-2 replication in HeLa-ACE2 cells at low micromolar IC<sub>50</sub>s. These studies suggest that TX and/or TP could possibly be repurposed for the prevention and/or treatment of SARS-CoV-2 infection.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2024-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11450544/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142381603","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}
Asmaa S A Yassen, Sherief M Abdel-Wahab, Khaled M Darwish, Mohamed S Nafie, Reda F A Abdelhameed, Gharieb S El-Sayyad, Ahmed I El-Batal, Khadiga M Attia, Hosam A Elshihawy, Ranza Elrayess
VEGFR2 inhibition has been established as a therapeutic approach for managing cancer. A series of curcumin-based analogues were designed, synthesized, and screened for their anticancer activity against MCF-7 and HepG-2 cell lines and WISH normal cells. Compounds 4b, 4d, 4e, and 4f showed potent cytotoxicity against MCF-7 with IC50 values of 0.49, 0.14, 0.01, and 0.32 μM, respectively, compared to curcumin (IC50 = 13.8 μM) and sorafenib (IC50 = 2.13 μM). Interestingly, compound 4e, the most active compound, exhibited potent VEGFR2 inhibition with an IC50 value of 11.6 nM (96.5% inhibition) compared to sorafenib with an IC50 value of 30 nM (94.8% inhibition). Additionally, compound 4e significantly induced apoptotic cell death in MCF-7 cells by 41.1% compared to a control group (0.8%), halting cell division during the G2/M phase by 39.8% compared to the control (21.7%). Molecular docking-coupled dynamics simulations highlighted the bias of the VEGFR2 pocket towards compound 4e compared to other synthesized compounds. Predicting superior binding affinities and relevant interactions with the pocket's key residues recapitulated in vitro findings towards higher inhibition activity for compound 4e. Furthermore, compound 4e with adequate pharmacokinetic and drug-likeness profiles in terms of ADME and safety characteristics can serve as a promising clinical candidate for future lead optimization and development. Notably, 4e-Fe2O3-humic acid NPs exhibited potent cytotoxicity with IC50 values of 2.41 and 13.4 ng mL-1 against MCF-7 and HepG-2 cell lines, respectively. Hence, compound 4e and its Fe2O3-humic acid-NPs could be further developed as promising anti-breast cancer agents.
抑制血管内皮生长因子受体 2 已被确定为治疗癌症的一种方法。我们设计、合成了一系列姜黄素类似物,并筛选了它们对 MCF-7 和 HepG-2 细胞系以及 WISH 正常细胞的抗癌活性。与姜黄素(IC50 = 13.8 μM)和索拉非尼(IC50 = 2.13 μM)相比,化合物 4b、4d、4e 和 4f 对 MCF-7 具有很强的细胞毒性,IC50 值分别为 0.49、0.14、0.01 和 0.32 μM。有趣的是,活性最高的化合物 4e 对血管内皮生长因子受体 2 具有强效抑制作用,其 IC50 值为 11.6 nM(抑制率为 96.5%),而索拉非尼的 IC50 值为 30 nM(抑制率为 94.8%)。此外,与对照组(0.8%)相比,化合物 4e 能显著诱导 MCF-7 细胞凋亡 41.1%;与对照组(21.7%)相比,化合物 4e 能使细胞在 G2/M 期停止分裂 39.8%。分子对接-耦合动力学模拟突出表明,与其他合成化合物相比,VEGFR2 口袋偏向于化合物 4e。通过预测化合物 4e 与口袋关键残基的卓越结合亲和力和相关相互作用,再现了体外研究结果,即化合物 4e 具有更高的抑制活性。此外,化合物 4e 在药代动力学和药物相似性方面具有充分的 ADME 和安全性特征,可作为有前途的临床候选化合物,用于未来的先导化合物优化和开发。值得注意的是,4e-Fe2O3-腐植酸 NPs 对 MCF-7 和 HepG-2 细胞株具有很强的细胞毒性,IC50 值分别为 2.41 和 13.4 ng mL-1。因此,化合物 4e 及其 Fe2O3-humic acid-NPs 可进一步开发为有前景的抗乳腺癌药物。
{"title":"Novel curcumin-based analogues as potential VEGFR2 inhibitors with promising metallic loading nanoparticles: synthesis, biological evaluation, and molecular modelling investigation.","authors":"Asmaa S A Yassen, Sherief M Abdel-Wahab, Khaled M Darwish, Mohamed S Nafie, Reda F A Abdelhameed, Gharieb S El-Sayyad, Ahmed I El-Batal, Khadiga M Attia, Hosam A Elshihawy, Ranza Elrayess","doi":"10.1039/d4md00574k","DOIUrl":"https://doi.org/10.1039/d4md00574k","url":null,"abstract":"<p><p>VEGFR2 inhibition has been established as a therapeutic approach for managing cancer. A series of curcumin-based analogues were designed, synthesized, and screened for their anticancer activity against MCF-7 and HepG-2 cell lines and WISH normal cells. Compounds 4b, 4d, 4e, and 4f showed potent cytotoxicity against MCF-7 with IC<sub>50</sub> values of 0.49, 0.14, 0.01, and 0.32 μM, respectively, compared to curcumin (IC<sub>50</sub> = 13.8 μM) and sorafenib (IC<sub>50</sub> = 2.13 μM). Interestingly, compound 4e, the most active compound, exhibited potent VEGFR2 inhibition with an IC<sub>50</sub> value of 11.6 nM (96.5% inhibition) compared to sorafenib with an IC<sub>50</sub> value of 30 nM (94.8% inhibition). Additionally, compound 4e significantly induced apoptotic cell death in MCF-7 cells by 41.1% compared to a control group (0.8%), halting cell division during the G2/M phase by 39.8% compared to the control (21.7%). Molecular docking-coupled dynamics simulations highlighted the bias of the VEGFR2 pocket towards compound 4e compared to other synthesized compounds. Predicting superior binding affinities and relevant interactions with the pocket's key residues recapitulated <i>in vitro</i> findings towards higher inhibition activity for compound 4e. Furthermore, compound 4e with adequate pharmacokinetic and drug-likeness profiles in terms of ADME and safety characteristics can serve as a promising clinical candidate for future lead optimization and development. Notably, 4e-Fe<sub>2</sub>O<sub>3</sub>-humic acid NPs exhibited potent cytotoxicity with IC<sub>50</sub> values of 2.41 and 13.4 ng mL<sup>-1</sup> against MCF-7 and HepG-2 cell lines, respectively. Hence, compound 4e and its Fe<sub>2</sub>O<sub>3</sub>-humic acid-NPs could be further developed as promising anti-breast cancer agents.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2024-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11428034/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142353066","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}
Tommaso Felicetti, Nicola Di Iacovo, Maria Agnese Della Fazia, Danilo Piobbico, Stefania Pieroni, Martina Pacetti, Jialing Yu, Yilun Sun, Serena Massari, Maria Letizia Barreca, Stefano Sabatini, Oriana Tabarrini, Violetta Cecchetti, Fei Wang, Yves Pommier, Mariangela Morlando, Giuseppe Servillo, Giuseppe Manfroni
MicroRNAs (miRNAs) play a crucial role in ovarian cancer (OC) pathogenesis and miRNA processing can be the object of pharmacological intervention. By exploiting our in-house quinolone library, we combined a cell-based screening with medicinal chemistry efforts, ultimately leading to derivative 33 with anti-OC activity against distinct cell lines (GI50 values 13.52-31.04 μM) and CC50 Wi-38 = 142.9 μM. Compound 33 retained anticancer activity against additional cancer cells and demonstrated a synergistic effect with cisplatin against cisplatin-resistant A2780 cells. Compound 33 bound TRBP by SPR (KD = 4.09 μM) and thermal shift assays and its activity was TRBP-dependent, leading to modulation of siRNA and miRNA maturation. Derivative 33 exhibited augmented potency against OC cells and a stronger binding affinity for TRBP compared to enoxacin, the sole quinolone identified as a modulator of miRNA maturation. Consequently, 33 represents a promising template for developing novel anti-OC agents with a distinctive mechanism of action.
{"title":"New anti-ovarian cancer quinolone derivatives acting by modulating microRNA processing machinery.","authors":"Tommaso Felicetti, Nicola Di Iacovo, Maria Agnese Della Fazia, Danilo Piobbico, Stefania Pieroni, Martina Pacetti, Jialing Yu, Yilun Sun, Serena Massari, Maria Letizia Barreca, Stefano Sabatini, Oriana Tabarrini, Violetta Cecchetti, Fei Wang, Yves Pommier, Mariangela Morlando, Giuseppe Servillo, Giuseppe Manfroni","doi":"10.1039/d4md00649f","DOIUrl":"https://doi.org/10.1039/d4md00649f","url":null,"abstract":"<p><p>MicroRNAs (miRNAs) play a crucial role in ovarian cancer (OC) pathogenesis and miRNA processing can be the object of pharmacological intervention. By exploiting our in-house quinolone library, we combined a cell-based screening with medicinal chemistry efforts, ultimately leading to derivative 33 with anti-OC activity against distinct cell lines (GI<sub>50</sub> values 13.52-31.04 μM) and CC<sub>50 Wi-38</sub> = 142.9 μM. Compound 33 retained anticancer activity against additional cancer cells and demonstrated a synergistic effect with cisplatin against cisplatin-resistant A2780 cells. Compound 33 bound TRBP by SPR (<i>K</i> <sub>D</sub> = 4.09 μM) and thermal shift assays and its activity was TRBP-dependent, leading to modulation of siRNA and miRNA maturation. Derivative 33 exhibited augmented potency against OC cells and a stronger binding affinity for TRBP compared to enoxacin, the sole quinolone identified as a modulator of miRNA maturation. Consequently, 33 represents a promising template for developing novel anti-OC agents with a distinctive mechanism of action.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2024-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11467779/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142473775","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}
Sofia Khabirova, Mikhail Menshikov-Tonyan, Gleb Aleshin, Anastasia Prikhodko, Daniil Kozlov, Evgeny Anokhin, Konstantin Babeshkin, Nikolay Titchenko, Anastasia Zubenko, Anna Shchukina, Yuri Fedorov, Stepan Kalmykov
The application of nanoparticles is promising for the purposes of nuclear medicine due to the possibilities of using them as vectors and transporters of radionuclides. In this study, we have successfully synthesised conjugates of CeO2 nanoparticles and azacrown ligands. Then, the radiolabelling conditions with radionuclides 65Zn, 44Sc and 207Bi were selected and the kinetic stability of the complexes in biologically significant media was evaluated. Optimum conditions for CeO2-APTES-L and CeO2-APTES-DOTA labelling were found: 0.1 g l-1 conjugate and 10-9 M metal cations at 90 °C for complexes with [65Zn]Zn2+, [44Sc]Sc3+ and [207Bi]Bi3+. CeO2-APTES-L-44Sc (radiochemical purity more than 90%) was stable in fetal bovine serum. The obtained results enabled us to choose the most promising complex for biomedical applications for carrying out in vitro and in vivo biodistribution research. Nanoceria and its derivative showed no obvious toxicity to human endothelial cells EA.hy926. Then, the in vivo stability of the studied scandium complex was demonstrated. Taken together, our studies show that functionalised cerium oxide nanoparticles lead to stable radiolabelled nanosystems that may be used for targeted drug delivery, diagnosis and treatment of oncological diseases.
{"title":"Assessing the biocompatibility and stability of CeO<sub>2</sub> nanoparticle conjugates with azacrowns for use as radiopharmaceuticals.","authors":"Sofia Khabirova, Mikhail Menshikov-Tonyan, Gleb Aleshin, Anastasia Prikhodko, Daniil Kozlov, Evgeny Anokhin, Konstantin Babeshkin, Nikolay Titchenko, Anastasia Zubenko, Anna Shchukina, Yuri Fedorov, Stepan Kalmykov","doi":"10.1039/d4md00515e","DOIUrl":"https://doi.org/10.1039/d4md00515e","url":null,"abstract":"<p><p>The application of nanoparticles is promising for the purposes of nuclear medicine due to the possibilities of using them as vectors and transporters of radionuclides. In this study, we have successfully synthesised conjugates of CeO<sub>2</sub> nanoparticles and azacrown ligands. Then, the radiolabelling conditions with radionuclides <sup>65</sup>Zn, <sup>44</sup>Sc and <sup>207</sup>Bi were selected and the kinetic stability of the complexes in biologically significant media was evaluated. Optimum conditions for CeO<sub>2</sub>-APTES-L and CeO<sub>2</sub>-APTES-DOTA labelling were found: 0.1 g l<sup>-1</sup> conjugate and 10<sup>-9</sup> M metal cations at 90 °C for complexes with [<sup>65</sup>Zn]Zn<sup>2+</sup>, [<sup>44</sup>Sc]Sc<sup>3+</sup> and [<sup>207</sup>Bi]Bi<sup>3+</sup>. CeO<sub>2</sub>-APTES-L-<sup>44</sup>Sc (radiochemical purity more than 90%) was stable in fetal bovine serum. The obtained results enabled us to choose the most promising complex for biomedical applications for carrying out <i>in vitro</i> and <i>in vivo</i> biodistribution research. Nanoceria and its derivative showed no obvious toxicity to human endothelial cells EA.hy926. Then, the <i>in vivo</i> stability of the studied scandium complex was demonstrated. Taken together, our studies show that functionalised cerium oxide nanoparticles lead to stable radiolabelled nanosystems that may be used for targeted drug delivery, diagnosis and treatment of oncological diseases.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2024-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11428044/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142353060","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}
Laura P R Figueroa, Renato L de Carvalho, Renata G Almeida, Esther R S Paz, Emilay B T Diogo, Maria H Araujo, Warley S Borges, Victor F S Ramos, Rubem F S Menna-Barreto, James M Wood, John F Bower, Eufrânio N da Silva Júnior
The regioselective synthesis of functionalized naphthoquinones via the formation and capture of naphthoquinonynes has been used to prepare trypanocidal compounds. The target compounds are functionalized on the aromatic ring, leaving the quinoidal ring intact. Using this technique, eighteen functionalized naphthoquinones were succesfull obtained, divided in two main groups: the first scope using N-nucleophiles, and the second scope using pyridine N-oxides, with yields up to 74%. Evaluation against bloodstream trypomastigotes of T. cruzi has identified fourteen compounds that are more potent than benznidazole (Bz); for instance, compounds 29b-I and 30b, with IC50/24 h values of 10.5 and 10.1 μM, respectively, are approximately 10-fold more active than Bz. This study provides the first examples of the application of naphthoquinonyne chemistry for the synthesis of new compounds with potent trypanocidal activities.
{"title":"Generation and capture of naphthoquinonynes: a new frontier in the development of trypanocidal quinones <i>via</i> aryne chemistry.","authors":"Laura P R Figueroa, Renato L de Carvalho, Renata G Almeida, Esther R S Paz, Emilay B T Diogo, Maria H Araujo, Warley S Borges, Victor F S Ramos, Rubem F S Menna-Barreto, James M Wood, John F Bower, Eufrânio N da Silva Júnior","doi":"10.1039/d4md00558a","DOIUrl":"https://doi.org/10.1039/d4md00558a","url":null,"abstract":"<p><p>The regioselective synthesis of functionalized naphthoquinones <i>via</i> the formation and capture of naphthoquinonynes has been used to prepare trypanocidal compounds. The target compounds are functionalized on the aromatic ring, leaving the quinoidal ring intact. Using this technique, eighteen functionalized naphthoquinones were succesfull obtained, divided in two main groups: the first scope using <i>N</i>-nucleophiles, and the second scope using pyridine <i>N</i>-oxides, with yields up to 74%. Evaluation against bloodstream trypomastigotes of <i>T. cruzi</i> has identified fourteen compounds that are more potent than benznidazole (Bz); for instance, compounds 29b-I and 30b, with IC<sub>50</sub>/24 h values of 10.5 and 10.1 μM, respectively, are approximately 10-fold more active than Bz. This study provides the first examples of the application of naphthoquinonyne chemistry for the synthesis of new compounds with potent trypanocidal activities.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11539365/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142606194","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}