Small molecules targeting activating mutations within the epidermal growth factor receptor (EGFR) are efficacious anticancer agents, particularly in non-small cell lung cancer (NSCLC). Among these, lazertinib, a third-generation tyrosine kinase inhibitor (TKI), has recently gained FDA approval for use in combination with amivantamab, a dual EGFR/MET-targeting monoclonal antibody. This review delves into the discovery and development of lazertinib underscoring the improvements in medicinal chemistry properties, especially in comparison with osimertinib. Analysis of its structure-activity relationships (SAR), as outlined in the patent literature, reveals the structural diversity explored enroute to the candidate molecule. The resulting structure of lazertinib is distinguished among other TKIs due to the combination of the hydrophobic phenyl and hydrophilic amine substituents on the pyrazole. The structural basis for the selectivity against the T790M mutation is enabled by the substituted pyrazole moiety, which facilitates both van der Waals and H-bonding interactions with the EGFR kinase domain. Insights from this case study offer lessons that can inform the future design of kinase inhibitors with improved safety and efficacy profiles for cancer treatment and other diseases.
{"title":"Lazertinib: breaking the mold of third-generation EGFR inhibitors.","authors":"Kishan B Patel, David E Heppner","doi":"10.1039/d4md00800f","DOIUrl":"10.1039/d4md00800f","url":null,"abstract":"<p><p>Small molecules targeting activating mutations within the epidermal growth factor receptor (EGFR) are efficacious anticancer agents, particularly in non-small cell lung cancer (NSCLC). Among these, lazertinib, a third-generation tyrosine kinase inhibitor (TKI), has recently gained FDA approval for use in combination with amivantamab, a dual EGFR/MET-targeting monoclonal antibody. This review delves into the discovery and development of lazertinib underscoring the improvements in medicinal chemistry properties, especially in comparison with osimertinib. Analysis of its structure-activity relationships (SAR), as outlined in the patent literature, reveals the structural diversity explored enroute to the candidate molecule. The resulting structure of lazertinib is distinguished among other TKIs due to the combination of the hydrophobic phenyl and hydrophilic amine substituents on the pyrazole. The structural basis for the selectivity against the T790M mutation is enabled by the substituted pyrazole moiety, which facilitates both van der Waals and H-bonding interactions with the EGFR kinase domain. Insights from this case study offer lessons that can inform the future design of kinase inhibitors with improved safety and efficacy profiles for cancer treatment and other diseases.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11758113/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143047313","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}
Timaeus E F Morgan, Emma K Grant, Robert C Shaw, Lachlan J N Waddell, Martyn C Henry, Holly McErlain, Carlos J Alcaide-Corral, Sally L Pimlott, Adriana A S Tavares, Andrew Sutherland
The sphingosine-1-phosphate-5 (S1P5) receptor is one of the five membrane G protein-coupled receptors that are activated by the lysophospholipid, sphingosine-1-phosphate, resulting in regulation of many cellular processes. S1P5 receptors are located on oligodendrocytes and are proposed to influence oligodendrocyte physiology. Understanding S1P5 modulation during processes such as remyelination could have potential applications for demyelinating CNS disorders such as multiple sclerosis (MS). Herein, we report the synthesis and preliminary evaluation of a series of fluorinated 6-arylaminobenzamides as positron emission tomography (PET) ligands of S1P5. Pharmacokinetic screening and binding evaluation using a [35S]GTPγS assay led to the discovery of TEFM78, a selective and high affinity agonist of S1P5. Radiosynthesis of [18F]TEFM78 allowed pilot PET imaging studies in an animal model, which showed that [18F]TEFM78 can cross the blood brain barrier with good uptake in rat brain and spinal cord.
{"title":"Synthesis and evaluation of 6-arylaminobenzamides as positron emission tomography imaging ligands for the sphingosine-1-phosphate-5 receptor.","authors":"Timaeus E F Morgan, Emma K Grant, Robert C Shaw, Lachlan J N Waddell, Martyn C Henry, Holly McErlain, Carlos J Alcaide-Corral, Sally L Pimlott, Adriana A S Tavares, Andrew Sutherland","doi":"10.1039/d4md00929k","DOIUrl":"10.1039/d4md00929k","url":null,"abstract":"<p><p>The sphingosine-1-phosphate-5 (S1P<sub>5</sub>) receptor is one of the five membrane G protein-coupled receptors that are activated by the lysophospholipid, sphingosine-1-phosphate, resulting in regulation of many cellular processes. S1P<sub>5</sub> receptors are located on oligodendrocytes and are proposed to influence oligodendrocyte physiology. Understanding S1P<sub>5</sub> modulation during processes such as remyelination could have potential applications for demyelinating CNS disorders such as multiple sclerosis (MS). Herein, we report the synthesis and preliminary evaluation of a series of fluorinated 6-arylaminobenzamides as positron emission tomography (PET) ligands of S1P<sub>5</sub>. Pharmacokinetic screening and binding evaluation using a [<sup>35</sup>S]GTPγS assay led to the discovery of TEFM78, a selective and high affinity agonist of S1P<sub>5</sub>. Radiosynthesis of [<sup>18</sup>F]TEFM78 allowed pilot PET imaging studies in an animal model, which showed that [<sup>18</sup>F]TEFM78 can cross the blood brain barrier with good uptake in rat brain and spinal cord.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2025-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11729640/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143010733","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}
Topical delivery of therapeutics on the skin can effectively alleviate skin symptoms of psoriasis and reduce systemic toxicity. However, the low delivery efficiency caused by the stratum corneum barrier limits the therapeutic impact. Here, we reported an oligopeptide hydrogel that encapsulates cell-penetrating-peptide (CPP)-decorated curcumin-loaded nanoemulsions (Cur-CNEs) to enhance the skin penetration of curcumin for topical treatment of psoriasis. After being applied to the skin of psoriatic mice, the Cur-CNE embedded oligopeptide hydrogel (Cur-CNEs/Gel) provided a prolonged residue time of Cur-CNEs on the skin lesion. The fluidic and elastic properties of the nanoemulsions enabled them to effectively pass through the interstitial spaces of the stratum corneum, while the CPP decoration further enhanced skin penetration and cellular uptake of Cur-CNEs. The Cur-CNEs/Gel exhibits effective alleviation of the symptoms of psoriasis in mice and provides a promising strategy for topical treatment of psoriasis.
{"title":"Enhanced skin penetration of curcumin by a nanoemulsion-embedded oligopeptide hydrogel for psoriasis topical therapy.","authors":"Kehan Chen, Hui Yang, Guo Xu, Yunhan Hu, Xue Tian, Song Qin, Tianyue Jiang","doi":"10.1039/d4md00781f","DOIUrl":"https://doi.org/10.1039/d4md00781f","url":null,"abstract":"<p><p>Topical delivery of therapeutics on the skin can effectively alleviate skin symptoms of psoriasis and reduce systemic toxicity. However, the low delivery efficiency caused by the stratum corneum barrier limits the therapeutic impact. Here, we reported an oligopeptide hydrogel that encapsulates cell-penetrating-peptide (CPP)-decorated curcumin-loaded nanoemulsions (Cur-CNEs) to enhance the skin penetration of curcumin for topical treatment of psoriasis. After being applied to the skin of psoriatic mice, the Cur-CNE embedded oligopeptide hydrogel (Cur-CNEs/Gel) provided a prolonged residue time of Cur-CNEs on the skin lesion. The fluidic and elastic properties of the nanoemulsions enabled them to effectively pass through the interstitial spaces of the stratum corneum, while the CPP decoration further enhanced skin penetration and cellular uptake of Cur-CNEs. The Cur-CNEs/Gel exhibits effective alleviation of the symptoms of psoriasis in mice and provides a promising strategy for topical treatment of psoriasis.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11694649/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142932563","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}
Diana McDonough, Christian A Sanchez, William M Wuest, Kevin P C Minbiole
The development and characterization of quaternary phosphonium compounds (QPCs) have long benefitted from their incorporation into a cornerstone reaction in organic synthesis - the Wittig reaction. These structures have, more recently, been developed into a wide variety of novel applications, ranging from phase transfer catalysis to mitochondrial targeting. Importantly, their antimicrobial action has demonstrated great promise against a wide variety of bacteria. This review aims to provide an overview of recent development in non-polymeric biocidal QPC structures, highlighting their synthetic preparation, and comparing their antimicrobial performance. Discussions of similarities and dissimilarities to QACs are included, both in bioactivity as well as likely mechanism(s) of action. The observed potential of QPCs to eradicate Gram-negative pathogens via a novel mechanism is highlighted, as there is an urgent need to address the declining biocide arsenal in modern infection control.
{"title":"Recent developments in antimicrobial small molecule quaternary phosphonium compounds (QPCs) - synthesis and biological insights.","authors":"Diana McDonough, Christian A Sanchez, William M Wuest, Kevin P C Minbiole","doi":"10.1039/d4md00855c","DOIUrl":"10.1039/d4md00855c","url":null,"abstract":"<p><p>The development and characterization of quaternary phosphonium compounds (QPCs) have long benefitted from their incorporation into a cornerstone reaction in organic synthesis - the Wittig reaction. These structures have, more recently, been developed into a wide variety of novel applications, ranging from phase transfer catalysis to mitochondrial targeting. Importantly, their antimicrobial action has demonstrated great promise against a wide variety of bacteria. This review aims to provide an overview of recent development in non-polymeric biocidal QPC structures, highlighting their synthetic preparation, and comparing their antimicrobial performance. Discussions of similarities and dissimilarities to QACs are included, both in bioactivity as well as likely mechanism(s) of action. The observed potential of QPCs to eradicate Gram-negative pathogens <i>via</i> a novel mechanism is highlighted, as there is an urgent need to address the declining biocide arsenal in modern infection control.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11729670/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143010645","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}
Shaun W P Rees, Tayla A Rees, Emily K Paulin, Olivia R Arnerich, Euphemia Leung, Christopher S Walker, David Barker, Lisa I Pilkington
Dysregulation of choline phospholipid metabolism and overexpression of phosphatidylcholine-specific phospholipase C (PC-PLC) is implicated in various cancers. Current known enzyme inhibitors include compounds based on a 2-morpholino-5-N-benzylamino benzoic acid, or hydroxamic acid, scaffold. In this work, 81 compounds were made by modifying this core structure to explore the pharmacophore. Specifically, these novel compounds result from changes to the central ring substitution pattern, alkyl heterocycle and methylation of the N-benzyl bridge. The anti-proliferative activity of the synthesised compounds was assessed against cancer cell lines MDA-MB-231 and HCT116. PC-PLCBC enzyme inhibition was also assessed, and the development of a pharmacokinetic profile was initiated using a microsomal stability assay. The findings confirmed the optimal pharmacophore as a 2-morpholino-5-N-benzylamino benzoic acid, or acid derivative, scaffold, and that this family of molecules demonstrate a high degree of stability following treatment with rat microsomes. Additionally, benzylic N-methylated compounds were the most biologically active compounds, encouraging further investigation into this region of the pharmacophore.
胆碱磷脂代谢失调和磷脂酰胆碱特异性磷脂酶C (PC-PLC)的过度表达与多种癌症有关。目前已知的酶抑制剂包括基于2-morpholino-5- n -苄基苯甲酸或羟肟酸支架的化合物。本研究通过对该核心结构进行修饰,合成了81个药效团化合物。具体来说,这些新化合物是由中心环取代模式、烷基杂环和n -苄基桥甲基化的变化引起的。合成的化合物对肿瘤细胞株MDA-MB-231和HCT116的抗增殖活性进行了评价。还评估了PC-PLCBC酶抑制,并使用微粒体稳定性测定法开始了药代动力学谱的开发。研究结果证实,最佳药效团是2-morpholino-5-N-benzylamino苯甲酸,或酸衍生物,支架,并且该分子家族在大鼠微粒体处理后表现出高度的稳定性。此外,苯基n甲基化化合物是最具生物活性的化合物,鼓励进一步研究药效团的这一区域。
{"title":"Structure-activity relationship expansion and microsomal stability assessment of the 2-morpholinobenzoic acid scaffold as antiproliferative phosphatidylcholine-specific phospholipase C inhibitors.","authors":"Shaun W P Rees, Tayla A Rees, Emily K Paulin, Olivia R Arnerich, Euphemia Leung, Christopher S Walker, David Barker, Lisa I Pilkington","doi":"10.1039/d4md00831f","DOIUrl":"10.1039/d4md00831f","url":null,"abstract":"<p><p>Dysregulation of choline phospholipid metabolism and overexpression of phosphatidylcholine-specific phospholipase C (PC-PLC) is implicated in various cancers. Current known enzyme inhibitors include compounds based on a 2-morpholino-5-<i>N</i>-benzylamino benzoic acid, or hydroxamic acid, scaffold. In this work, 81 compounds were made by modifying this core structure to explore the pharmacophore. Specifically, these novel compounds result from changes to the central ring substitution pattern, alkyl heterocycle and methylation of the <i>N</i>-benzyl bridge. The anti-proliferative activity of the synthesised compounds was assessed against cancer cell lines MDA-MB-231 and HCT116. PC-PLC<sub>BC</sub> enzyme inhibition was also assessed, and the development of a pharmacokinetic profile was initiated using a microsomal stability assay. The findings confirmed the optimal pharmacophore as a 2-morpholino-5-<i>N</i>-benzylamino benzoic acid, or acid derivative, scaffold, and that this family of molecules demonstrate a high degree of stability following treatment with rat microsomes. Additionally, benzylic <i>N</i>-methylated compounds were the most biologically active compounds, encouraging further investigation into this region of the pharmacophore.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11734694/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143010728","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}
Degrons are short amino acid sequences that can facilitate the degradation of protein substrates. They can be classified as either ubiquitin-dependent or -independent based on their interactions with the ubiquitin proteasome system (UPS). These amino acid sequences are often found in exposed regions of proteins serving as either a tethering point for an interaction with an E3 ligase or initiating signaling for the direct degradation of the protein. Recent advancements in the protein degradation field have shown the therapeutic potential of both classes of degrons through leveraging their degradative effects to engage specific protein targets. This review explores what targeted protein degradation applications degrons can be used in and how they have inspired new degrader technology to target a wide variety of protein substrates.
{"title":"Exploration of degrons and their ability to mediate targeted protein degradation.","authors":"Timothy J Harris, Darci J Trader","doi":"10.1039/d4md00787e","DOIUrl":"10.1039/d4md00787e","url":null,"abstract":"<p><p>Degrons are short amino acid sequences that can facilitate the degradation of protein substrates. They can be classified as either ubiquitin-dependent or -independent based on their interactions with the ubiquitin proteasome system (UPS). These amino acid sequences are often found in exposed regions of proteins serving as either a tethering point for an interaction with an E3 ligase or initiating signaling for the direct degradation of the protein. Recent advancements in the protein degradation field have shown the therapeutic potential of both classes of degrons through leveraging their degradative effects to engage specific protein targets. This review explores what targeted protein degradation applications degrons can be used in and how they have inspired new degrader technology to target a wide variety of protein substrates.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11758578/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143047841","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}
Aberrant protein misfolding and accumulation is considered to be a major pathological pillar of neurodegenerative disorders, including Alzheimer's and Parkinson's diseases. Aggregation of amyloid-β (Aβ) peptide leads to the formation of toxic amyloid fibrils and is associated with cognitive dysfunction and memory loss in Alzheimer's disease (AD). Designing molecules that inhibit amyloid aggregation seems to be a rational approach to AD drug development. Over the years, researchers have utilized a variety of therapeutic strategies targeting different pathways, extensively studying peptide-based approaches to understand AD pathology and demonstrate their efficacy against Aβ aggregation. This review highlights rationally designed peptide/mimetics, including structure-based peptides, metal-peptide chelators, stapled peptides, and peptide-based nanomaterials as potential amyloid inhibitors.
{"title":"Peptide-based amyloid-beta aggregation inhibitors.","authors":"Naina Sehra, Rajesh Parmar, Rahul Jain","doi":"10.1039/d4md00729h","DOIUrl":"https://doi.org/10.1039/d4md00729h","url":null,"abstract":"<p><p>Aberrant protein misfolding and accumulation is considered to be a major pathological pillar of neurodegenerative disorders, including Alzheimer's and Parkinson's diseases. Aggregation of amyloid-β (Aβ) peptide leads to the formation of toxic amyloid fibrils and is associated with cognitive dysfunction and memory loss in Alzheimer's disease (AD). Designing molecules that inhibit amyloid aggregation seems to be a rational approach to AD drug development. Over the years, researchers have utilized a variety of therapeutic strategies targeting different pathways, extensively studying peptide-based approaches to understand AD pathology and demonstrate their efficacy against Aβ aggregation. This review highlights rationally designed peptide/mimetics, including structure-based peptides, metal-peptide chelators, stapled peptides, and peptide-based nanomaterials as potential amyloid inhibitors.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11773382/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143067504","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}
Rouming Peng, Xin Liu, Chun-Chi Chen, Rey-Ting Guo, Jian Min
Despite the success of endocrine therapies in treating ER-positive breast cancer, the development of resistance remains a significant challenge. Estrogen receptor targeting proteolysis-targeting chimeras (ER PROTACs) offer a unique approach by harnessing the ubiquitin-proteasome system to degrade ER, potentially bypassing resistance mechanisms. In this review, we present the drug design, efficacy and early clinical trials of these ER PROTACs. This review underscores the academic and industrial opportunities presented by this emerging technology, as well as the challenges that must be addressed to translate these findings into effective clinical therapies.
{"title":"Development of PROTACs targeting estrogen receptor: an emerging technique for combating endocrine resistance.","authors":"Rouming Peng, Xin Liu, Chun-Chi Chen, Rey-Ting Guo, Jian Min","doi":"10.1039/d4md00961d","DOIUrl":"10.1039/d4md00961d","url":null,"abstract":"<p><p>Despite the success of endocrine therapies in treating ER-positive breast cancer, the development of resistance remains a significant challenge. Estrogen receptor targeting proteolysis-targeting chimeras (ER PROTACs) offer a unique approach by harnessing the ubiquitin-proteasome system to degrade ER, potentially bypassing resistance mechanisms. In this review, we present the drug design, efficacy and early clinical trials of these ER PROTACs. This review underscores the academic and industrial opportunities presented by this emerging technology, as well as the challenges that must be addressed to translate these findings into effective clinical therapies.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2024-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11734508/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143010528","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}
Sapna Saini, G Lakshma Reddy, Anjali Gangwar, Harpreet Kour, Gajanan G Nadre, Ramajayan Pandian, Sunny Pal, Utpal Nandi, Rashmi Sharma, Sanghapal D Sawant
Nitrofuran and pyrazolopyrimidine-based compounds possess a broad antimicrobial spectrum including Gram-positive and Gram-negative bacteria. In the present work, a series of conjugates of these scaffolds was synthesized and evaluated for antimicrobial activity against Staphylococcus aureus and methicillin-resistant S. aureus (MRSA). Many compounds showed MIC values of ≤2 μg ml-1, with compound 35 demonstrating excellent activity (MICs: 0.7 and 0.15 μg ml-1 against S. aureus and MRSA, respectively) and safety up to 50 μg ml-1 in HepG2 cells. Compound 35 also exhibited no hemolytic activity, biofilm eradication, and effectiveness against efflux-pump-overexpressing strains (NorA, TetK, MsrA) without resistance development. It showed synergistic effects with vancomycin (S. aureus) and rifampicin (MRSA). Mechanistic studies revealed that compound 35 exhibits good membrane-targeting abilities, as evidenced by DAPI/PI staining and scanning electron microscopy (SEM). In an intracellular model, it reduced bacterial load efficiently in both S. aureus and MRSA strains. With a strong in vitro profile, compound 35 demonstrated favorable oral pharmacokinetics at 30 mg kg-1 and potent in vivo anti-MRSA activity, highlighting its potential against antibiotic-resistant infections.
{"title":"Discovery and biological evaluation of nitrofuranyl-pyrazolopyrimidine hybrid conjugates as potent antimicrobial agents targeting <i>Staphylococcus aureus</i> and methicillin-resistant <i>S. aureus</i>.","authors":"Sapna Saini, G Lakshma Reddy, Anjali Gangwar, Harpreet Kour, Gajanan G Nadre, Ramajayan Pandian, Sunny Pal, Utpal Nandi, Rashmi Sharma, Sanghapal D Sawant","doi":"10.1039/d4md00826j","DOIUrl":"10.1039/d4md00826j","url":null,"abstract":"<p><p>Nitrofuran and pyrazolopyrimidine-based compounds possess a broad antimicrobial spectrum including Gram-positive and Gram-negative bacteria. In the present work, a series of conjugates of these scaffolds was synthesized and evaluated for antimicrobial activity against <i>Staphylococcus aureus</i> and methicillin-resistant <i>S. aureus</i> (MRSA). Many compounds showed MIC values of ≤2 μg ml<sup>-1</sup>, with compound 35 demonstrating excellent activity (MICs: 0.7 and 0.15 μg ml<sup>-1</sup> against <i>S. aureus</i> and MRSA, respectively) and safety up to 50 μg ml<sup>-1</sup> in HepG2 cells. Compound 35 also exhibited no hemolytic activity, biofilm eradication, and effectiveness against efflux-pump-overexpressing strains (NorA, TetK, MsrA) without resistance development. It showed synergistic effects with vancomycin (<i>S. aureus</i>) and rifampicin (MRSA). Mechanistic studies revealed that compound 35 exhibits good membrane-targeting abilities, as evidenced by DAPI/PI staining and scanning electron microscopy (SEM). In an intracellular model, it reduced bacterial load efficiently in both <i>S. aureus</i> and MRSA strains. With a strong <i>in vitro</i> profile, compound 35 demonstrated favorable oral pharmacokinetics at 30 mg kg<sup>-1</sup> and potent <i>in vivo</i> anti-MRSA activity, highlighting its potential against antibiotic-resistant infections.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2024-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11740095/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143010448","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}
Rebecca Stevens, Harry J Shrives, Jenni Cryan, Diana Klimaszewska, Peter Stacey, Glenn A Burley, John D Harling, David J Battersby, Afjal H Miah
High-throughput chemistry (HTC) and direct-to-biology (D2B) platforms allow for plate-based compound synthesis and biological evaluation of crude mixtures in cellular assays. The rise of these workflows has rapidly accelerated drug-discovery programs in the field of targeted protein degradation (TPD) in recent years by removing a key bottleneck of compound purification. However, the number of chemical transformations amenable to this methodology remain minimal, leading to limitations in the exploration of chemical space using existing library-based approaches. In this work, we expanded the toolbox by synthesising a library of degraders in D2B format. First, reaction conditions are established for performing key medicinal chemistry transformations, including reductive amination, SNAr, palladium-mediated cross-coupling and alkylation, in D2B format. Second, the utility of these alternative reactions is demonstrated by rapidly identifying developable PROTACs for a range of protein targets.
{"title":"Expanding the reaction toolbox for nanoscale direct-to-biology PROTAC synthesis and biological evaluation.","authors":"Rebecca Stevens, Harry J Shrives, Jenni Cryan, Diana Klimaszewska, Peter Stacey, Glenn A Burley, John D Harling, David J Battersby, Afjal H Miah","doi":"10.1039/d4md00760c","DOIUrl":"10.1039/d4md00760c","url":null,"abstract":"<p><p>High-throughput chemistry (HTC) and direct-to-biology (D2B) platforms allow for plate-based compound synthesis and biological evaluation of crude mixtures in cellular assays. The rise of these workflows has rapidly accelerated drug-discovery programs in the field of targeted protein degradation (TPD) in recent years by removing a key bottleneck of compound purification. However, the number of chemical transformations amenable to this methodology remain minimal, leading to limitations in the exploration of chemical space using existing library-based approaches. In this work, we expanded the toolbox by synthesising a library of degraders in D2B format. First, reaction conditions are established for performing key medicinal chemistry transformations, including reductive amination, S<sub>N</sub>Ar, palladium-mediated cross-coupling and alkylation, in D2B format. Second, the utility of these alternative reactions is demonstrated by rapidly identifying developable PROTACs for a range of protein targets.</p>","PeriodicalId":21462,"journal":{"name":"RSC medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2024-12-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11664481/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142885684","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}