Zosurabalpin:一种能杀死耐碳青霉烯类鲍曼不动杆菌的新型大环肽抗生素。

IF 10.7 Q1 MEDICINE, RESEARCH & EXPERIMENTAL MedComm Pub Date : 2024-08-11 DOI:10.1002/mco2.696
Qi Weng, Feng Zhang, Quan Zheng
{"title":"Zosurabalpin:一种能杀死耐碳青霉烯类鲍曼不动杆菌的新型大环肽抗生素。","authors":"Qi Weng,&nbsp;Feng Zhang,&nbsp;Quan Zheng","doi":"10.1002/mco2.696","DOIUrl":null,"url":null,"abstract":"<p>In two recent studies published back-to-back in <i>Nature</i>, Bradley's and Kahne's teams reported a novel tethered macrocyclic peptide (MCP) antibiotic called zosurabalpin, which targets the LptB2FGC complex in the inner membrane to block lipopolysaccharide (LPS) transport, leading to the accumulation of this endotoxin in the cell, ultimately resulting in the death of the bacteria (Figure 1).<span><sup>1, 2</sup></span> Zosurabalpin not only showed excellent antibacterial activity against carbapenem-resistant <i>Acinetobacter baumannii</i> (CRAB) in vivo and in vitro, but is also expected to break through the resistance mechanisms of existing antibiotics, offering new prospects for clinical treatment.</p><p><i>A. baumannii</i>, a Gram-negative bacterium with an outer membrane containing LPS, is resistant to penetration by a wide range of antibiotics, making it difficult to kill. It is one of the common causative agents of hospital-acquired infections and results in hospital-acquired pneumonia, bloodstream infections, urinary tract infections, and soft tissue skin infections. In recent years, with the extensive use of antibiotics, the resistance of <i>A. baumannii</i> has been increasing, bringing great challenges to clinical treatment. One of the infections caused by CRAB is particularly difficult to treat, which is listed as a priority on the World Health Organization's list of key pathogens and also as an urgent threat by the Centers for Disease Control and Prevention in the United States.<span><sup>3</sup></span> Therefore, the development of novel antibiotics against Gram-negative bacteria has become an urgent research topic for researchers.</p><p>In order to find novel antibiotics against CRAB, Bradley's team screened and optimized 44,985 MCPs produced by Tranzyme Pharma for antimicrobial activity (both Gram-negative and Gram-positive), and identified the leading compound, RO7075573, which showed significant inhibition of <i>A. baumannii</i> with a minimum inhibitory concentration (MIC) of 0.12 mg L<sup>−1</sup>.<span><sup>1</sup></span> It also demonstrated high selectivity and was ineffective against wild-type, exocytosis-impaired, and pore-protein-deficient <i>Escherichia coli</i>, <i>Klebsiella pneumoniae</i>, and <i>Pseudomonas aeruginosa</i>. In addition, RO7075573 showed similar antimicrobial activity against antibiotic-sensitive strains and multidrug-resistant <i>A. baumannii</i> strains, suggesting that its mechanism of action is different from that of the current clinically applied antibiotics and that it may overcome existing resistance mechanisms. However, severe tolerance issues were observed in rat models of intravenous administration. Therefore, the researchers optimized the structure of the MCPs, and successfully acquired an amphoteric benzoic acid derivative, zosurabalpin. It showed an MIC of 0.25 mg L<sup>−1</sup> against <i>A. baumannii</i>, and demonstrated superior tolerability.</p><p>To identify potential molecular targets of zosurabalpin, <i>A. baumannii</i> was induced to spontaneously develop resistance by incrementally increasing the concentration of zosurabalpin in the medium, which was then analyzed by gene sequencing to reveal mutation sites. The researchers found 28 differential mutations in the gene encoding LptF and two unique mutations in LptG. As LptF and LptG are components of the LptB2FGC complex, which is a key part of the LPS transport system, this suggests that zosurabalpin may target the LptB2FGC complex to block LPS transport. In addition, zosurabalpin not only showed good pharmacokinetics with high clearance (51 mL min<sup>−1</sup> kg<sup>−1</sup>), low volume of distribution (0.7 L kg<sup>−1</sup>), short terminal half-life (0.3 h), and moderate protein binding (fraction unbound, 37%), but also demonstrated good in vivo efficacy in mouse models of infection, including sepsis and femur and lung infections caused by CRAB strains. Furthermore, phase I clinical studies have demonstrated that a single intravenous dose of 10–2000 mg of zosurabalpin is generally safe and well tolerated.<span><sup>4</sup></span></p><p>Meanwhile, another study elucidated for the mechanism of the antibacterial activity of tethered MCP antibiotics (including zosurabalpin, RO7196472 and RO7075573).<span><sup>2</sup></span> First, Kahne's team observed by cryo-electron microscopy (cryo-EM) that RO7196472 binds to LptB2FG as well as LPS to form a ternary complex, in which RO7196472 binds to pockets formed by the arrangement of the side chains of several amino acids in the transmembrane (TM) helices of LptF (Glu58, Glu249, Trp271, Val314, Ile317, Arg320, and Thr321) and LptG (Leu36). More importantly, Kahne's team also observed that RO7196472 captures an intermediate state of LptB2FG–LPS. However, the overall conformation of LptB2FG–LPS remained essentially stable with or without RO7196472, suggesting that the cyclic peptide binds to the pre-existing structure of the LptB2FG–LPS complex, thus confirming that this state is a druggable conformation for antibiotic development.</p><p>LptC is a member of the LptB2FGC complex, and plays an important role in the transfer of LPS from LptF to LptA (Figure 1A).<span><sup>5</sup></span> By analyzing the cryo-EM structures of zosurabalpin, RO7196472, and RO7075573 bound to the LptB2FG–LPS complex while comparing the efficacy of these three MCP antibiotics, Kahne's team found that the MCP-targeted LptC TM helix had dissociated from the complex (Figure 1B). In addition, the results of ATPase activity and LPS release assays further validated that the compounds had the strongest affinity for LptB2FG–LPS in the absence of LptC. However, the researchers found that <i>A. baylyi</i> strains lacking LPS in the outer membrane could grow in vitro in the presence of high concentrations of RO7196472, suggesting that MCPs do not act by depleting LPS from the outer membrane, but rather by its toxic accumulation within the cell. As LPS is a key component of the outer membrane of Gram-negative bacteria, its abnormal intracellular accumulation may interfere with normal intracellular physiological activities, including enzyme activity, the conduct of metabolic pathways, and cell signaling. When LPS accumulates to a certain level, it may trigger intracellular death procedures, such as activation of autolysin or induction of apoptosis, leading to cell death. Therefore, it is necessary to further explore the specific mechanism by which zosurabalpin contributes to the intracellular accumulation of LPS, and thus induces cell death. On the one hand, we can analyze the changes in gene expression and protein expression in zosurabalpin-treated bacteria to identify the molecular pathways involved in response to LPS accumulation. On the other hand, we can investigate the effects of LPS accumulation on bacterial metabolic pathways, especially those related to energy production, amino acid synthesis, and lipid metabolism.</p><p>In summary, zosurabalpin, with its distinctive mechanism of action, selective antibacterial activity, highly potent in vitro and in vivo efficacy, and favorable tolerability, exhibits notable competitive advantages in the domain of CRAB therapy. First, zosurabalpin is highly selective for CRAB and has limited activity against other Gram-negative and Gram-positive bacteria, helping to minimize the impact on normal flora and thus potentially reducing associated side effects. Second, due to the novel mechanism of action of zosurabalpin, CRAB currently lacks an effective defense mechanism in the face of zosurabalpin, which will provide a new option for the clinical treatment of drug-resistant strains. However, researchers have found that selective mutations in the LPS transport mechanism can reduce the potency of the drug, implying that <i>Acinetobacter</i> strains will inevitably develop resistance to this class of antibiotics as well. Consequently, it is imperative to investigate the potential of combining zosurabalpin with other antibiotics to enhance therapeutic efficacy and impede the emergence of resistance. Meanwhile, it is essential to investigate the mechanisms by which Gram-negative bacteria develop resistance to zosurabalpin, as well as the means of preventing and reversing such resistance. In addition, the LPS transporter LptB2FGC complex, as a new potential antibiotic target, provides ideas for the future development and design of synthetic novel antibiotics. We can also further chemically modify MCPs to improve their stability, bioavailability, and reduce potential side effects.</p><p><b>Quan Zheng</b> conceived the manuscript. <b>Qi Weng</b> and <b>Quan Zheng</b> wrote the manuscript. <b>Qi Weng</b> prepared the figure. <b>Feng Zhang</b> proofread and edited the manuscript. All authors have read and approved the article.</p><p>The authors declare they have no conflicts of interest.</p><p>Not applicable.</p>","PeriodicalId":94133,"journal":{"name":"MedComm","volume":null,"pages":null},"PeriodicalIF":10.7000,"publicationDate":"2024-08-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11317178/pdf/","citationCount":"0","resultStr":"{\"title\":\"Zosurabalpin: a novel tethered macrocyclic peptide antibiotic that kills carbapenem-resistant Acinetobacter baumannii\",\"authors\":\"Qi Weng,&nbsp;Feng Zhang,&nbsp;Quan Zheng\",\"doi\":\"10.1002/mco2.696\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>In two recent studies published back-to-back in <i>Nature</i>, Bradley's and Kahne's teams reported a novel tethered macrocyclic peptide (MCP) antibiotic called zosurabalpin, which targets the LptB2FGC complex in the inner membrane to block lipopolysaccharide (LPS) transport, leading to the accumulation of this endotoxin in the cell, ultimately resulting in the death of the bacteria (Figure 1).<span><sup>1, 2</sup></span> Zosurabalpin not only showed excellent antibacterial activity against carbapenem-resistant <i>Acinetobacter baumannii</i> (CRAB) in vivo and in vitro, but is also expected to break through the resistance mechanisms of existing antibiotics, offering new prospects for clinical treatment.</p><p><i>A. baumannii</i>, a Gram-negative bacterium with an outer membrane containing LPS, is resistant to penetration by a wide range of antibiotics, making it difficult to kill. It is one of the common causative agents of hospital-acquired infections and results in hospital-acquired pneumonia, bloodstream infections, urinary tract infections, and soft tissue skin infections. In recent years, with the extensive use of antibiotics, the resistance of <i>A. baumannii</i> has been increasing, bringing great challenges to clinical treatment. One of the infections caused by CRAB is particularly difficult to treat, which is listed as a priority on the World Health Organization's list of key pathogens and also as an urgent threat by the Centers for Disease Control and Prevention in the United States.<span><sup>3</sup></span> Therefore, the development of novel antibiotics against Gram-negative bacteria has become an urgent research topic for researchers.</p><p>In order to find novel antibiotics against CRAB, Bradley's team screened and optimized 44,985 MCPs produced by Tranzyme Pharma for antimicrobial activity (both Gram-negative and Gram-positive), and identified the leading compound, RO7075573, which showed significant inhibition of <i>A. baumannii</i> with a minimum inhibitory concentration (MIC) of 0.12 mg L<sup>−1</sup>.<span><sup>1</sup></span> It also demonstrated high selectivity and was ineffective against wild-type, exocytosis-impaired, and pore-protein-deficient <i>Escherichia coli</i>, <i>Klebsiella pneumoniae</i>, and <i>Pseudomonas aeruginosa</i>. In addition, RO7075573 showed similar antimicrobial activity against antibiotic-sensitive strains and multidrug-resistant <i>A. baumannii</i> strains, suggesting that its mechanism of action is different from that of the current clinically applied antibiotics and that it may overcome existing resistance mechanisms. However, severe tolerance issues were observed in rat models of intravenous administration. Therefore, the researchers optimized the structure of the MCPs, and successfully acquired an amphoteric benzoic acid derivative, zosurabalpin. It showed an MIC of 0.25 mg L<sup>−1</sup> against <i>A. baumannii</i>, and demonstrated superior tolerability.</p><p>To identify potential molecular targets of zosurabalpin, <i>A. baumannii</i> was induced to spontaneously develop resistance by incrementally increasing the concentration of zosurabalpin in the medium, which was then analyzed by gene sequencing to reveal mutation sites. The researchers found 28 differential mutations in the gene encoding LptF and two unique mutations in LptG. As LptF and LptG are components of the LptB2FGC complex, which is a key part of the LPS transport system, this suggests that zosurabalpin may target the LptB2FGC complex to block LPS transport. In addition, zosurabalpin not only showed good pharmacokinetics with high clearance (51 mL min<sup>−1</sup> kg<sup>−1</sup>), low volume of distribution (0.7 L kg<sup>−1</sup>), short terminal half-life (0.3 h), and moderate protein binding (fraction unbound, 37%), but also demonstrated good in vivo efficacy in mouse models of infection, including sepsis and femur and lung infections caused by CRAB strains. Furthermore, phase I clinical studies have demonstrated that a single intravenous dose of 10–2000 mg of zosurabalpin is generally safe and well tolerated.<span><sup>4</sup></span></p><p>Meanwhile, another study elucidated for the mechanism of the antibacterial activity of tethered MCP antibiotics (including zosurabalpin, RO7196472 and RO7075573).<span><sup>2</sup></span> First, Kahne's team observed by cryo-electron microscopy (cryo-EM) that RO7196472 binds to LptB2FG as well as LPS to form a ternary complex, in which RO7196472 binds to pockets formed by the arrangement of the side chains of several amino acids in the transmembrane (TM) helices of LptF (Glu58, Glu249, Trp271, Val314, Ile317, Arg320, and Thr321) and LptG (Leu36). More importantly, Kahne's team also observed that RO7196472 captures an intermediate state of LptB2FG–LPS. However, the overall conformation of LptB2FG–LPS remained essentially stable with or without RO7196472, suggesting that the cyclic peptide binds to the pre-existing structure of the LptB2FG–LPS complex, thus confirming that this state is a druggable conformation for antibiotic development.</p><p>LptC is a member of the LptB2FGC complex, and plays an important role in the transfer of LPS from LptF to LptA (Figure 1A).<span><sup>5</sup></span> By analyzing the cryo-EM structures of zosurabalpin, RO7196472, and RO7075573 bound to the LptB2FG–LPS complex while comparing the efficacy of these three MCP antibiotics, Kahne's team found that the MCP-targeted LptC TM helix had dissociated from the complex (Figure 1B). In addition, the results of ATPase activity and LPS release assays further validated that the compounds had the strongest affinity for LptB2FG–LPS in the absence of LptC. However, the researchers found that <i>A. baylyi</i> strains lacking LPS in the outer membrane could grow in vitro in the presence of high concentrations of RO7196472, suggesting that MCPs do not act by depleting LPS from the outer membrane, but rather by its toxic accumulation within the cell. As LPS is a key component of the outer membrane of Gram-negative bacteria, its abnormal intracellular accumulation may interfere with normal intracellular physiological activities, including enzyme activity, the conduct of metabolic pathways, and cell signaling. When LPS accumulates to a certain level, it may trigger intracellular death procedures, such as activation of autolysin or induction of apoptosis, leading to cell death. Therefore, it is necessary to further explore the specific mechanism by which zosurabalpin contributes to the intracellular accumulation of LPS, and thus induces cell death. On the one hand, we can analyze the changes in gene expression and protein expression in zosurabalpin-treated bacteria to identify the molecular pathways involved in response to LPS accumulation. On the other hand, we can investigate the effects of LPS accumulation on bacterial metabolic pathways, especially those related to energy production, amino acid synthesis, and lipid metabolism.</p><p>In summary, zosurabalpin, with its distinctive mechanism of action, selective antibacterial activity, highly potent in vitro and in vivo efficacy, and favorable tolerability, exhibits notable competitive advantages in the domain of CRAB therapy. First, zosurabalpin is highly selective for CRAB and has limited activity against other Gram-negative and Gram-positive bacteria, helping to minimize the impact on normal flora and thus potentially reducing associated side effects. Second, due to the novel mechanism of action of zosurabalpin, CRAB currently lacks an effective defense mechanism in the face of zosurabalpin, which will provide a new option for the clinical treatment of drug-resistant strains. However, researchers have found that selective mutations in the LPS transport mechanism can reduce the potency of the drug, implying that <i>Acinetobacter</i> strains will inevitably develop resistance to this class of antibiotics as well. Consequently, it is imperative to investigate the potential of combining zosurabalpin with other antibiotics to enhance therapeutic efficacy and impede the emergence of resistance. Meanwhile, it is essential to investigate the mechanisms by which Gram-negative bacteria develop resistance to zosurabalpin, as well as the means of preventing and reversing such resistance. In addition, the LPS transporter LptB2FGC complex, as a new potential antibiotic target, provides ideas for the future development and design of synthetic novel antibiotics. We can also further chemically modify MCPs to improve their stability, bioavailability, and reduce potential side effects.</p><p><b>Quan Zheng</b> conceived the manuscript. <b>Qi Weng</b> and <b>Quan Zheng</b> wrote the manuscript. <b>Qi Weng</b> prepared the figure. <b>Feng Zhang</b> proofread and edited the manuscript. All authors have read and approved the article.</p><p>The authors declare they have no conflicts of interest.</p><p>Not applicable.</p>\",\"PeriodicalId\":94133,\"journal\":{\"name\":\"MedComm\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":10.7000,\"publicationDate\":\"2024-08-11\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11317178/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"MedComm\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/mco2.696\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"MEDICINE, RESEARCH & EXPERIMENTAL\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"MedComm","FirstCategoryId":"1085","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/mco2.696","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"MEDICINE, RESEARCH & EXPERIMENTAL","Score":null,"Total":0}
引用次数: 0

摘要

5 通过分析与 LptB2FG-LPS 复合物结合的 zosurabalpin、RO7196472 和 RO7075573 的冷冻电镜结构,同时比较这三种 MCP 抗生素的功效,Kahne 的团队发现 MCP 靶向的 LptC TM 螺旋已经从复合物中解离(图 1B)。此外,ATPase 活性和 LPS 释放试验的结果进一步验证了这些化合物在没有 LptC 的情况下对 LptB2FG-LPS 的亲和力最强。然而,研究人员发现,外膜中缺乏 LPS 的 A. baylyi 菌株可以在高浓度 RO7196472 的存在下体外生长,这表明 MCPs 并不是通过消耗外膜中的 LPS 而发挥作用,而是通过其在细胞内的毒性积累而发挥作用。由于 LPS 是革兰氏阴性细菌外膜的关键成分,其在细胞内的异常积累可能会干扰细胞内的正常生理活动,包括酶活性、代谢途径的进行和细胞信号的传递。当 LPS 积累到一定水平时,可能会触发细胞内的死亡程序,如激活自溶酶或诱导细胞凋亡,从而导致细胞死亡。因此,有必要进一步探讨佐舒拉巴平促进 LPS 在细胞内蓄积,从而诱导细胞死亡的具体机制。一方面,我们可以分析经唑舒巴平处理的细菌中基因表达和蛋白质表达的变化,以确定参与应对 LPS 积累的分子通路。总之,佐舒拉巴平具有独特的作用机制、选择性抗菌活性、强效的体外和体内疗效以及良好的耐受性,在 CRAB 治疗领域具有显著的竞争优势。首先,Zosurabalpin 对 CRAB 具有高度选择性,对其他革兰氏阴性菌和革兰氏阳性菌的活性有限,有助于最大限度地减少对正常菌群的影响,从而可能减少相关副作用。其次,由于 zosurabalpin 的作用机制新颖,CRAB 目前面对 zosurabalpin 缺乏有效的防御机制,这将为耐药菌株的临床治疗提供新的选择。然而,研究人员发现,LPS 转运机制的选择性突变会降低药物的效力,这意味着不动杆菌菌株也将不可避免地对这一类抗生素产生耐药性。因此,当务之急是研究佐舒拉巴平与其他抗生素联用的可能性,以提高疗效并阻止耐药性的产生。同时,研究革兰氏阴性细菌对唑喹恶啉产生耐药性的机制,以及预防和逆转这种耐药性的方法也至关重要。此外,LPS 转运体 LptB2FGC 复合物作为一个新的潜在抗生素靶点,为未来合成新型抗生素的开发和设计提供了思路。我们还可以进一步对MCPs进行化学修饰,以提高其稳定性、生物利用度并减少潜在的副作用。翁琦和郑权撰写了手稿。翁琦绘制了图表。张锋校对并编辑了手稿。所有作者均已阅读并认可该文章。作者声明他们没有利益冲突。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

摘要图片

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
Zosurabalpin: a novel tethered macrocyclic peptide antibiotic that kills carbapenem-resistant Acinetobacter baumannii

In two recent studies published back-to-back in Nature, Bradley's and Kahne's teams reported a novel tethered macrocyclic peptide (MCP) antibiotic called zosurabalpin, which targets the LptB2FGC complex in the inner membrane to block lipopolysaccharide (LPS) transport, leading to the accumulation of this endotoxin in the cell, ultimately resulting in the death of the bacteria (Figure 1).1, 2 Zosurabalpin not only showed excellent antibacterial activity against carbapenem-resistant Acinetobacter baumannii (CRAB) in vivo and in vitro, but is also expected to break through the resistance mechanisms of existing antibiotics, offering new prospects for clinical treatment.

A. baumannii, a Gram-negative bacterium with an outer membrane containing LPS, is resistant to penetration by a wide range of antibiotics, making it difficult to kill. It is one of the common causative agents of hospital-acquired infections and results in hospital-acquired pneumonia, bloodstream infections, urinary tract infections, and soft tissue skin infections. In recent years, with the extensive use of antibiotics, the resistance of A. baumannii has been increasing, bringing great challenges to clinical treatment. One of the infections caused by CRAB is particularly difficult to treat, which is listed as a priority on the World Health Organization's list of key pathogens and also as an urgent threat by the Centers for Disease Control and Prevention in the United States.3 Therefore, the development of novel antibiotics against Gram-negative bacteria has become an urgent research topic for researchers.

In order to find novel antibiotics against CRAB, Bradley's team screened and optimized 44,985 MCPs produced by Tranzyme Pharma for antimicrobial activity (both Gram-negative and Gram-positive), and identified the leading compound, RO7075573, which showed significant inhibition of A. baumannii with a minimum inhibitory concentration (MIC) of 0.12 mg L−1.1 It also demonstrated high selectivity and was ineffective against wild-type, exocytosis-impaired, and pore-protein-deficient Escherichia coli, Klebsiella pneumoniae, and Pseudomonas aeruginosa. In addition, RO7075573 showed similar antimicrobial activity against antibiotic-sensitive strains and multidrug-resistant A. baumannii strains, suggesting that its mechanism of action is different from that of the current clinically applied antibiotics and that it may overcome existing resistance mechanisms. However, severe tolerance issues were observed in rat models of intravenous administration. Therefore, the researchers optimized the structure of the MCPs, and successfully acquired an amphoteric benzoic acid derivative, zosurabalpin. It showed an MIC of 0.25 mg L−1 against A. baumannii, and demonstrated superior tolerability.

To identify potential molecular targets of zosurabalpin, A. baumannii was induced to spontaneously develop resistance by incrementally increasing the concentration of zosurabalpin in the medium, which was then analyzed by gene sequencing to reveal mutation sites. The researchers found 28 differential mutations in the gene encoding LptF and two unique mutations in LptG. As LptF and LptG are components of the LptB2FGC complex, which is a key part of the LPS transport system, this suggests that zosurabalpin may target the LptB2FGC complex to block LPS transport. In addition, zosurabalpin not only showed good pharmacokinetics with high clearance (51 mL min−1 kg−1), low volume of distribution (0.7 L kg−1), short terminal half-life (0.3 h), and moderate protein binding (fraction unbound, 37%), but also demonstrated good in vivo efficacy in mouse models of infection, including sepsis and femur and lung infections caused by CRAB strains. Furthermore, phase I clinical studies have demonstrated that a single intravenous dose of 10–2000 mg of zosurabalpin is generally safe and well tolerated.4

Meanwhile, another study elucidated for the mechanism of the antibacterial activity of tethered MCP antibiotics (including zosurabalpin, RO7196472 and RO7075573).2 First, Kahne's team observed by cryo-electron microscopy (cryo-EM) that RO7196472 binds to LptB2FG as well as LPS to form a ternary complex, in which RO7196472 binds to pockets formed by the arrangement of the side chains of several amino acids in the transmembrane (TM) helices of LptF (Glu58, Glu249, Trp271, Val314, Ile317, Arg320, and Thr321) and LptG (Leu36). More importantly, Kahne's team also observed that RO7196472 captures an intermediate state of LptB2FG–LPS. However, the overall conformation of LptB2FG–LPS remained essentially stable with or without RO7196472, suggesting that the cyclic peptide binds to the pre-existing structure of the LptB2FG–LPS complex, thus confirming that this state is a druggable conformation for antibiotic development.

LptC is a member of the LptB2FGC complex, and plays an important role in the transfer of LPS from LptF to LptA (Figure 1A).5 By analyzing the cryo-EM structures of zosurabalpin, RO7196472, and RO7075573 bound to the LptB2FG–LPS complex while comparing the efficacy of these three MCP antibiotics, Kahne's team found that the MCP-targeted LptC TM helix had dissociated from the complex (Figure 1B). In addition, the results of ATPase activity and LPS release assays further validated that the compounds had the strongest affinity for LptB2FG–LPS in the absence of LptC. However, the researchers found that A. baylyi strains lacking LPS in the outer membrane could grow in vitro in the presence of high concentrations of RO7196472, suggesting that MCPs do not act by depleting LPS from the outer membrane, but rather by its toxic accumulation within the cell. As LPS is a key component of the outer membrane of Gram-negative bacteria, its abnormal intracellular accumulation may interfere with normal intracellular physiological activities, including enzyme activity, the conduct of metabolic pathways, and cell signaling. When LPS accumulates to a certain level, it may trigger intracellular death procedures, such as activation of autolysin or induction of apoptosis, leading to cell death. Therefore, it is necessary to further explore the specific mechanism by which zosurabalpin contributes to the intracellular accumulation of LPS, and thus induces cell death. On the one hand, we can analyze the changes in gene expression and protein expression in zosurabalpin-treated bacteria to identify the molecular pathways involved in response to LPS accumulation. On the other hand, we can investigate the effects of LPS accumulation on bacterial metabolic pathways, especially those related to energy production, amino acid synthesis, and lipid metabolism.

In summary, zosurabalpin, with its distinctive mechanism of action, selective antibacterial activity, highly potent in vitro and in vivo efficacy, and favorable tolerability, exhibits notable competitive advantages in the domain of CRAB therapy. First, zosurabalpin is highly selective for CRAB and has limited activity against other Gram-negative and Gram-positive bacteria, helping to minimize the impact on normal flora and thus potentially reducing associated side effects. Second, due to the novel mechanism of action of zosurabalpin, CRAB currently lacks an effective defense mechanism in the face of zosurabalpin, which will provide a new option for the clinical treatment of drug-resistant strains. However, researchers have found that selective mutations in the LPS transport mechanism can reduce the potency of the drug, implying that Acinetobacter strains will inevitably develop resistance to this class of antibiotics as well. Consequently, it is imperative to investigate the potential of combining zosurabalpin with other antibiotics to enhance therapeutic efficacy and impede the emergence of resistance. Meanwhile, it is essential to investigate the mechanisms by which Gram-negative bacteria develop resistance to zosurabalpin, as well as the means of preventing and reversing such resistance. In addition, the LPS transporter LptB2FGC complex, as a new potential antibiotic target, provides ideas for the future development and design of synthetic novel antibiotics. We can also further chemically modify MCPs to improve their stability, bioavailability, and reduce potential side effects.

Quan Zheng conceived the manuscript. Qi Weng and Quan Zheng wrote the manuscript. Qi Weng prepared the figure. Feng Zhang proofread and edited the manuscript. All authors have read and approved the article.

The authors declare they have no conflicts of interest.

Not applicable.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
CiteScore
6.70
自引率
0.00%
发文量
0
审稿时长
10 weeks
期刊最新文献
Copper homeostasis and cuproptosis in health and disease Liver cirrhosis: molecular mechanisms and therapeutic interventions High serum magnesium level is associated with increased mortality in patients with sepsis: an international, multicenter retrospective study N1-methylpseudouridine modification level correlates with protein expression, immunogenicity, and stability of mRNA Issue Information
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1