首页 > 最新文献

Antimicrobial Agents and Chemotherapy最新文献

英文 中文
In vitro PK/PD modeling and simulation to accurately assess the antimicrobial activity of tigecycline against Mycobacterium abscessus. 通过体外PK/PD建模和模拟,准确评估替加环素对脓肿分枝杆菌的抗菌活性。
IF 4.5 2区 医学 Q2 MICROBIOLOGY Pub Date : 2025-12-23 DOI: 10.1128/aac.01025-25
Hyunseo Park, Sara E Maloney Norcross, Anthony J Hickey, Mercedes Gonzalez-Juarrero, Bernd Meibohm

Conventional in vitro susceptibility testing methods may underestimate the bactericidal activity of antibiotics that are chemically unstable in aqueous media, thereby limiting their clinical translatability. Tigecycline is considered a representative example of such compounds, exhibiting notable therapeutic efficacy against a broad range of pathogens despite poor in vitro susceptibility profiles, as reflected by elevated MIC values. This discrepancy is likely attributable, at least in part, to the chemical instability of TGC under standard MIC assay conditions. In this manuscript, we propose a mechanism-based PK/PD modeling approach as a framework to overcome the limitations of traditional MIC assessments and to address potential discrepancies between intrinsic and experimentally measured apparent antibacterial activity. Dynamic time-kill curves for single and multiple dose scenarios of TGC against Mycobacterium abscessus (Mab) were experimentally simulated in 24-well plates, leveraging the chemical instability of TGC. Based on the resulting in vitro data, a mechanism-based model was developed to perform simulations for characterizing intrinsic efficacy and potency of TGC. While the in vitro MIC of TGC determined under standard conditions was determined as 3.125 mg/L, an intrinsic MIC simulated based on model predicted bacterial time-time kill curves was 0.5 mg/L. Model-based analysis also revealed that MIC under standard conditions was stemming from drug instability and bacterial growth rate in the utilized media. In conclusion, the PK/PD modeling and simulation-based MIC determination indicated that clinically achievable exposure levels of TGC are sufficient to kill Mab, underlining the therapeutic potential of TGC against Mab infections.

传统的体外药敏试验方法可能低估了抗生素在水介质中化学不稳定的杀菌活性,从而限制了它们的临床可翻译性。替加环素被认为是此类化合物的代表性例子,尽管体外敏感性较差,但对多种病原体表现出显着的治疗效果,这反映在MIC值升高上。这种差异可能至少部分归因于TGC在标准MIC测定条件下的化学不稳定性。在本文中,我们提出了一种基于机制的PK/PD建模方法作为框架,以克服传统MIC评估的局限性,并解决内在和实验测量的表观抗菌活性之间的潜在差异。利用TGC的化学不稳定性,在24孔板上模拟了单剂量和多剂量TGC对脓分枝杆菌(Mycobacterium abessus, Mab)的动态时间杀伤曲线。在体外实验数据的基础上,建立了一个基于机制的模型来模拟TGC的内在功效和效力。标准条件下测定的TGC体外MIC为3.125 mg/L,根据模型预测的细菌时间-时间杀伤曲线模拟的内禀MIC为0.5 mg/L。基于模型的分析还显示,标准条件下的MIC源于所用培养基中的药物不稳定性和细菌生长速度。总之,基于PK/PD模型和模拟的MIC测定表明,临床可达到的TGC暴露水平足以杀死单抗,强调了TGC对单抗感染的治疗潜力。
{"title":"<i>In vitro</i> PK/PD modeling and simulation to accurately assess the antimicrobial activity of tigecycline against <i>Mycobacterium abscessus</i>.","authors":"Hyunseo Park, Sara E Maloney Norcross, Anthony J Hickey, Mercedes Gonzalez-Juarrero, Bernd Meibohm","doi":"10.1128/aac.01025-25","DOIUrl":"https://doi.org/10.1128/aac.01025-25","url":null,"abstract":"<p><p>Conventional <i>in vitro</i> susceptibility testing methods may underestimate the bactericidal activity of antibiotics that are chemically unstable in aqueous media, thereby limiting their clinical translatability. Tigecycline is considered a representative example of such compounds, exhibiting notable therapeutic efficacy against a broad range of pathogens despite poor <i>in vitro</i> susceptibility profiles, as reflected by elevated MIC values. This discrepancy is likely attributable, at least in part, to the chemical instability of TGC under standard MIC assay conditions. In this manuscript, we propose a mechanism-based PK/PD modeling approach as a framework to overcome the limitations of traditional MIC assessments and to address potential discrepancies between intrinsic and experimentally measured apparent antibacterial activity. Dynamic time-kill curves for single and multiple dose scenarios of TGC against <i>Mycobacterium abscessus (Mab</i>) were experimentally simulated in 24-well plates, leveraging the chemical instability of TGC. Based on the resulting <i>in vitro</i> data, a mechanism-based model was developed to perform simulations for characterizing intrinsic efficacy and potency of TGC. While the <i>in vitro</i> MIC of TGC determined under standard conditions was determined as 3.125 mg/L, an intrinsic MIC simulated based on model predicted bacterial time-time kill curves was 0.5 mg/L. Model-based analysis also revealed that MIC under standard conditions was stemming from drug instability and bacterial growth rate in the utilized media. In conclusion, the PK/PD modeling and simulation-based MIC determination indicated that clinically achievable exposure levels of TGC are sufficient to kill <i>Mab</i>, underlining the therapeutic potential of TGC against <i>Mab</i> infections.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0102525"},"PeriodicalIF":4.5,"publicationDate":"2025-12-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145809180","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Antibacterial activity of enmetazobactam against Acinetobacter spp.: a molecular dissection of mechanism of action and resistance determinants. 恩美唑巴坦对不动杆菌的抑菌活性:作用机制和耐药决定因素的分子解剖。
IF 4.5 2区 医学 Q2 MICROBIOLOGY Pub Date : 2025-12-22 DOI: 10.1128/aac.01206-25
Gabriela-Alejandra Báez-Barroso, Arianna Rodríguez-Coello, Juan Carlos Vázquez-Ucha, Silvia López-Argüello, Michelle Outeda-García, Lucía González-Pinto, Andrea García-Pose, Paula Guijarro-Sánchez, Isaac Alonso-García, Emilio Lence, Concepción González-Bello, Antonio Oliver, Jorge Arca-Suárez, Bartolome Moya, Germán Bou, Alejandro Beceiro

The persistence of multidrug-resistant Acinetobacter baumannii remains a clinical challenge. Cefepime/enmetazobactam is a novel combination with demonstrated activity against extended-spectrum β-lactamase-producing Enterobacterales, but its activity against Acinetobacter has not yet been thoroughly explored. We aimed to assess its activity against Acinetobacter spp., including multidrug-resistant strains producing carbapenem-hydrolyzing class D β-lactamases (CHDLs). We analyzed 208 clinical isolates of Acinetobacter spp., including 67 carbapenem-resistant Acinetobacter baumannii (CRAB). Antibiotic susceptibility testing was conducted with cefepime, sulbactam, and imipenem, alone and in combination with enmetazobactam; the latter was also tested individually. Additionally, MICs of enmetazobactam/durlobactam and sulbactam/durlobactam were determined for CRAB and CHDL-producing A. baumannii ATCC 17978 transformants. PBP binding assays (IC₅₀), molecular docking, simulation studies with the enmetazobactam/OXA-23 adduct, hydrolysis kinetics (kcat, Km), and OXA-23 inhibition assays (IC₅₀, koff, t₁/₂) were performed to elucidate the mechanism of enmetazobactam and detect reduced susceptibility. Enmetazobactam showed high intrinsic activity against Acinetobacter spp., displaying reduced MICs against carbapenem-susceptible isolates. MIC50/90 of the enmetazobactam/durlobactam combination was 2/2 mg/L for CHDL-producing A. baumannii. Enmetazobactam exhibited bactericidal activity comparable to sulbactam. Binding assays revealed that the antimicrobial activity is driven by selective affinity for PBP2 (IC₅₀ 3.6 mg/L) and PBP3 (IC₅₀ 4.2 mg/L). OXA-23 readily inactivated enmetazobactam, confirming the major role of CHDLs in resistance to enmetazobactam, via substrate-assisted de-acylation. This study evidences the potent antimicrobial activity of enmetazobactam against A. baumannii via inhibition of PBP2 and PBP3. Its combination with new OXA-type inhibitors (e.g., durlobactam) represents a potential therapeutic alternative for multidrug-resistant A. baumannii.

耐多药鲍曼不动杆菌的持续存在仍然是一个临床挑战。头孢吡肟/恩美唑巴坦是一种新型组合,具有抗广谱β-内酰胺酶产肠杆菌的活性,但其抗不动杆菌的活性尚未得到充分的研究。我们的目的是评估其对不动杆菌的活性,包括产生碳青霉烯水解D类β-内酰胺酶(chdl)的多重耐药菌株。我们分析了208株临床分离的不动杆菌,包括67株耐碳青霉烯鲍曼不动杆菌(CRAB)。头孢吡肟、舒巴坦和亚胺培南单独或与恩美唑巴坦联合进行抗生素敏感性试验;后者也分别进行了测试。此外,还测定了恩美唑巴坦/杜氯巴坦和舒巴坦/杜氯巴坦对CRAB和产chdl鲍曼不动杆菌ATCC 17978转化体的mic。进行了PBP结合试验(IC₅0),分子对接,与enmetazobactam/OXA-23加合物的模拟研究,水解动力学(kcat, Km)和OXA-23抑制试验(IC₅0,koff, t₁/ 2),以阐明enmetazobactam的机制并检测降低的敏感性。恩美唑巴坦对不动杆菌表现出较高的内在活性,对碳青霉烯敏感的菌株表现出较低的mic。恩美唑巴坦/杜氯巴坦联合用药对产chdl鲍曼不动杆菌的MIC50/90为2/ 2mg /L。恩美唑巴坦表现出与舒巴坦相当的杀菌活性。结合实验显示,抗菌活性是由PBP2 (IC₅₀3.6 mg/L)和PBP3 (IC₅₀4.2 mg/L)的选择性亲和力驱动的。OXA-23很容易灭活恩美他唑巴坦,证实了chdl在恩美他唑巴坦耐药中的主要作用,通过底物辅助去酰化。本研究通过对PBP2和PBP3的抑制,证明了恩美唑巴坦对鲍曼不动杆菌具有较强的抑菌活性。它与新的oxa型抑制剂(如杜氯巴坦)联合使用,代表了多药耐药鲍曼不动杆菌的潜在治疗选择。
{"title":"Antibacterial activity of enmetazobactam against <i>Acinetobacter</i> spp.: a molecular dissection of mechanism of action and resistance determinants.","authors":"Gabriela-Alejandra Báez-Barroso, Arianna Rodríguez-Coello, Juan Carlos Vázquez-Ucha, Silvia López-Argüello, Michelle Outeda-García, Lucía González-Pinto, Andrea García-Pose, Paula Guijarro-Sánchez, Isaac Alonso-García, Emilio Lence, Concepción González-Bello, Antonio Oliver, Jorge Arca-Suárez, Bartolome Moya, Germán Bou, Alejandro Beceiro","doi":"10.1128/aac.01206-25","DOIUrl":"https://doi.org/10.1128/aac.01206-25","url":null,"abstract":"<p><p>The persistence of multidrug-resistant <i>Acinetobacter baumannii</i> remains a clinical challenge. Cefepime/enmetazobactam is a novel combination with demonstrated activity against extended-spectrum β-lactamase-producing Enterobacterales, but its activity against <i>Acinetobacter</i> has not yet been thoroughly explored. We aimed to assess its activity against <i>Acinetobacter</i> spp., including multidrug-resistant strains producing carbapenem-hydrolyzing class D β-lactamases (CHDLs). We analyzed 208 clinical isolates of <i>Acinetobacter</i> spp., including 67 carbapenem-resistant <i>Acinetobacter baumannii</i> (CRAB). Antibiotic susceptibility testing was conducted with cefepime, sulbactam, and imipenem, alone and in combination with enmetazobactam; the latter was also tested individually. Additionally, MICs of enmetazobactam/durlobactam and sulbactam/durlobactam were determined for CRAB and CHDL-producing <i>A. baumannii</i> ATCC 17978 transformants. PBP binding assays (IC₅₀), molecular docking, simulation studies with the enmetazobactam/OXA-23 adduct, hydrolysis kinetics (<i>k</i><sub>cat</sub>, <i>K</i><sub>m</sub>), and OXA-23 inhibition assays (IC₅₀, <i>k</i><sub>off</sub>, <i>t</i><sub>₁/₂</sub>) were performed to elucidate the mechanism of enmetazobactam and detect reduced susceptibility. Enmetazobactam showed high intrinsic activity against <i>Acinetobacter</i> spp., displaying reduced MICs against carbapenem-susceptible isolates. MIC<sub>50/90</sub> of the enmetazobactam/durlobactam combination was 2/2 mg/L for CHDL-producing <i>A. baumannii</i>. Enmetazobactam exhibited bactericidal activity comparable to sulbactam. Binding assays revealed that the antimicrobial activity is driven by selective affinity for PBP2 (IC₅₀ 3.6 mg/L) and PBP3 (IC₅₀ 4.2 mg/L). OXA-23 readily inactivated enmetazobactam, confirming the major role of CHDLs in resistance to enmetazobactam, via substrate-assisted de-acylation. This study evidences the potent antimicrobial activity of enmetazobactam against <i>A. baumannii</i> via inhibition of PBP2 and PBP3. Its combination with new OXA-type inhibitors (e.g., durlobactam) represents a potential therapeutic alternative for multidrug-resistant <i>A. baumannii</i>.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0120625"},"PeriodicalIF":4.5,"publicationDate":"2025-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145803010","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A mechanism-based pharmacokinetic/pharmacodynamic analysis of polymyxin B-based combination therapy against carbapenem-resistant Klebsiella pneumoniae isolates with diverse phenotypic and genotypic resistance mechanisms. 多粘菌素b联合治疗碳青霉烯耐药肺炎克雷伯菌不同表型和基因型耐药机制的药代动力学/药效学分析
IF 4.5 2区 医学 Q2 MICROBIOLOGY Pub Date : 2025-12-19 DOI: 10.1128/aac.00782-25
Ramya Mahadevan, Estefany Garcia, Rajnikant Sharma, Hongqiang Qiu, Ahmed Elsheikh, Robert Parambi, Cely Saad Abboud, Fernando Pasteran, Maria Soledad Ramirez, Keith S Kaye, Robert A Bonomo, Gauri G Rao

Increased resistance to β-lactams/β-lactamase inhibitors by mutations in β-lactamase genes, porins, and efflux pumps complicates the management of carbapenem-resistant Klebsiella pneumoniae (CRKP). Polymyxin B (PMB)-based combination therapy is the best alternative treatment for middle and low-income countries that cannot access the latest medicines. It is crucial to know both phenotypic and genotypic characteristics of a pathogen to understand the killing effect of each drug and its combinations. Hence, our objective was to incorporate mechanistic insights gained from resistance mechanisms of each isolate to develop a mechanism-based pharmacokinetic/pharmacodynamic model. Six clinical CRKP isolates with diverse genotypic resistance expressing blaKPC, blaNDM, porin, and mgrB mutations were used for static concentration time kill (SCTK) assays to evaluate the rate and extent of killing by monotherapy, double and triple combinations using PMB (0.5-64 mg/L), meropenem (10-120 mg/L), and fosfomycin (75-500 mg/L). Isolate BRKP28 expressed non-functional MgrB (a regulatory protein) and high-level phenotypic resistance (PMB MIC: >128 mg/L). In line with the observed resistance, the model estimated that BRKP28 had a reduced maximum killing rate constant for PMB (3.61 h⁻¹) relative to other isolates. The mechanistic synergy of PMB, due to outer membrane disruption, was incorporated into three isolates with porin mutations. PMB demonstrated 83%-88% mechanistic synergy with meropenem and 81%-98% with fosfomycin. The model further estimated that a very low concentration of PMB (0.49-0.64 mg/L) was sufficient to achieve 50% of the maximum synergy. Simulations using population pharmacokinetic models showed that combination therapy of PMB (1 mg/kg q12h) and fosfomycin (8 g q8h) achieved >73% reduction in area under the bacterial load-versus-time curve across four isolates. The triple combination therapy achieved a 67.7% reduction in non-carbapenamase producing isolate. These findings demonstrates that a low PMB dosing regimen (1 mg/kg q12h) can produce synergistic effects in combination therapy and may be effective in managing infections caused by CRKP, including PMB resistant isolates.

β-内酰胺酶基因、孔蛋白和外排泵突变导致β-内酰胺/β-内酰胺酶抑制剂耐药性增加,使碳青霉烯耐药肺炎克雷伯菌(CRKP)的管理复杂化。以多粘菌素B (PMB)为基础的联合治疗是无法获得最新药物的中低收入国家的最佳替代治疗。了解病原体的表型和基因型特征对了解每种药物及其组合的杀伤效果至关重要。因此,我们的目标是结合从每种分离物的耐药机制中获得的机制见解,以建立基于机制的药代动力学/药效学模型。采用6株表达blaKPC、blaNDM、porin和mgrB突变的不同基因型耐药临床CRKP分离株进行静态浓度时间杀伤(SCTK)试验,评估PMB (0.5-64 mg/L)、美罗培南(10-120 mg/L)和磷霉素(75-500 mg/L)单药、双药和三联药的杀伤率和程度。分离物BRKP28表达无功能MgrB(一种调节蛋白)和高水平表型抗性(PMB MIC: >128 mg/L)。根据观察到的耐药性,该模型估计BRKP28对PMB的最大杀伤率常数(3.61 h⁻¹)相对于其他菌株更低。由于外膜破坏,PMB的机制协同作用被纳入三个具有孔蛋白突变的分离株。PMB与美罗培南的协同作用为83% ~ 88%,与磷霉素的协同作用为81% ~ 98%。该模型进一步估计,极低浓度的PMB (0.49-0.64 mg/L)足以实现50%的最大协同作用。利用群体药代动力学模型模拟表明,PMB (1 mg/kg q12h)和磷霉素(8 g q8h)联合治疗可使4株菌株的细菌负荷-时间曲线上的面积减少约73%。三联疗法使非碳青霉烯酶产生分离物减少67.7%。这些发现表明,低剂量PMB方案(1mg /kg q12h)可在联合治疗中产生协同效应,并可能有效管理由CRKP引起的感染,包括PMB耐药分离株。
{"title":"A mechanism-based pharmacokinetic/pharmacodynamic analysis of polymyxin B-based combination therapy against carbapenem-resistant <i>Klebsiella pneumoniae</i> isolates with diverse phenotypic and genotypic resistance mechanisms.","authors":"Ramya Mahadevan, Estefany Garcia, Rajnikant Sharma, Hongqiang Qiu, Ahmed Elsheikh, Robert Parambi, Cely Saad Abboud, Fernando Pasteran, Maria Soledad Ramirez, Keith S Kaye, Robert A Bonomo, Gauri G Rao","doi":"10.1128/aac.00782-25","DOIUrl":"https://doi.org/10.1128/aac.00782-25","url":null,"abstract":"<p><p>Increased resistance to β-lactams/β-lactamase inhibitors by mutations in β-lactamase genes, porins, and efflux pumps complicates the management of carbapenem-resistant <i>Klebsiella pneumoniae</i> (CRKP). Polymyxin B (PMB)-based combination therapy is the best alternative treatment for middle and low-income countries that cannot access the latest medicines. It is crucial to know both phenotypic and genotypic characteristics of a pathogen to understand the killing effect of each drug and its combinations. Hence, our objective was to incorporate mechanistic insights gained from resistance mechanisms of each isolate to develop a mechanism-based pharmacokinetic/pharmacodynamic model. Six clinical CRKP isolates with diverse genotypic resistance expressing <i>bla</i><sub>KPC</sub>, <i>bla</i><sub>NDM</sub>, porin, and mgrB mutations were used for static concentration time kill (SCTK) assays to evaluate the rate and extent of killing by monotherapy, double and triple combinations using PMB (0.5-64 mg/L), meropenem (10-120 mg/L), and fosfomycin (75-500 mg/L). Isolate BRKP28 expressed non-functional MgrB (a regulatory protein) and high-level phenotypic resistance (PMB MIC: >128 mg/L). In line with the observed resistance, the model estimated that BRKP28 had a reduced maximum killing rate constant for PMB (3.61 h⁻¹) relative to other isolates. The mechanistic synergy of PMB, due to outer membrane disruption, was incorporated into three isolates with porin mutations. PMB demonstrated 83%-88% mechanistic synergy with meropenem and 81%-98% with fosfomycin. The model further estimated that a very low concentration of PMB (0.49-0.64 mg/L) was sufficient to achieve 50% of the maximum synergy. Simulations using population pharmacokinetic models showed that combination therapy of PMB (1 mg/kg q12h) and fosfomycin (8 g q8h) achieved >73% reduction in area under the bacterial load-versus-time curve across four isolates. The triple combination therapy achieved a 67.7% reduction in non-carbapenamase producing isolate. These findings demonstrates that a low PMB dosing regimen (1 mg/kg q12h) can produce synergistic effects in combination therapy and may be effective in managing infections caused by CRKP, including PMB resistant isolates.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0078225"},"PeriodicalIF":4.5,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145793003","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Preclinical pharmacokinetics/pharmacodynamics studies defining the role of ethambutol in Mycobacterium kansasii lung disease. 确定乙胺丁醇在堪萨斯分枝杆菌肺病中的作用的临床前药代动力学/药效学研究。
IF 4.5 2区 医学 Q2 MICROBIOLOGY Pub Date : 2025-12-19 DOI: 10.1128/aac.01447-25
Tawanda Gumbo, Gunavanthi D Boorgula, Shashikant Srivastava

Rifampin, isoniazid, and ethambutol are the backbone of the regimen used to treat Mycobacterium kansasii-complex (MKC) lung disease. However, ethambutol pharmacokinetics/pharmacodynamics (PK/PD) studies to inform on optimal exposure target and clinical dose for MKC are lacking. We performed studies to determine ethambutol minimum inhibitory concentration (MIC), mutation frequency (3× MIC), a PK/PD study using the hollow fiber system model of MKC (HFS-MKC) using the reference ATCC#12478 strain, and Monte Carlo simulation experiments for clinical dose selection and susceptibility breakpoint. We also performed a literature search to generate ethambutol MIC distribution for MKC. First, nine studies were identified with MIC of 587 isolates, and MIC50 and MIC90 identified as 4 and 16 mg/L, respectively. Second, the ethambutol MIC of the ATCC strain was 8 mg/L, and the mutation frequency was 4.23 × 10-2 CFU/mL. Third, in the HFS-MKC, ethambutol failed to kill M. kansasii below stasis (B0), and resistance emerged rapidly. The target exposure was an AUC0-24/MIC of 5.47 (95% confidence interval: 1.17-9.77). Fourth, Monte Carlo experiments of 10,000 virtual subjects identified doses of 1,200 and 3,000 mg to achieve or exceed target exposure in 18.21% and 58.57% of patients; and PK/PD MIC susceptibility breakpoints were determined as 2 and 4 mg/L, respectively. Doses >1,200 mg/day may have a higher likelihood of ocular toxicity. The risk of toxicity versus no microbial kill benefit in HFS-MKC suggests the need for better drugs compared to ethambutol in the treatment of MKC lung disease.

利福平、异烟肼和乙胺丁醇是治疗堪萨斯分枝杆菌复合体(MKC)肺部疾病的主要药物。然而,目前尚缺乏乙胺丁醇药代动力学/药效学(PK/PD)研究来确定MKC的最佳暴露靶点和临床剂量。我们进行了乙胺丁醇最低抑制浓度(MIC)、突变频率(3× MIC)的测定研究,以参考菌株ATCC#12478为MKC中空纤维系统模型(HFS-MKC)进行了PK/PD研究,并进行了临床剂量选择和药敏断点的蒙特卡罗模拟实验。我们还进行了文献检索,以生成MKC的乙胺丁醇MIC分布。首先,对9个研究的587株菌株进行MIC鉴定,MIC50和MIC90分别鉴定为4和16 mg/L。其次,ATCC菌株的乙胺丁醇MIC为8 mg/L,突变频率为4.23 × 10-2 CFU/mL。第三,在HFS-MKC中,乙胺丁醇未能杀死处于停滞期(B0)以下的堪萨斯结核分枝杆菌,并迅速产生耐药性。目标暴露AUC0-24/MIC为5.47(95%可信区间:1.17-9.77)。第四,1万名虚拟受试者的蒙特卡罗实验发现,18.21%和58.57%的患者在1200和3000 mg剂量下达到或超过目标暴露;PK/PD MIC药敏断点分别为2和4 mg/L。剂量低于1200mg /天可能有更高的眼毒性。HFS-MKC的毒性风险与无微生物杀灭益处的风险表明,在治疗MKC肺部疾病时,需要比乙胺丁醇更好的药物。
{"title":"Preclinical pharmacokinetics/pharmacodynamics studies defining the role of ethambutol in <i>Mycobacterium kansasii</i> lung disease.","authors":"Tawanda Gumbo, Gunavanthi D Boorgula, Shashikant Srivastava","doi":"10.1128/aac.01447-25","DOIUrl":"https://doi.org/10.1128/aac.01447-25","url":null,"abstract":"<p><p>Rifampin, isoniazid, and ethambutol are the backbone of the regimen used to treat <i>Mycobacterium kansasii-</i>complex (MKC) lung disease. However, ethambutol pharmacokinetics/pharmacodynamics (PK/PD) studies to inform on optimal exposure target and clinical dose for MKC are lacking. We performed studies to determine ethambutol minimum inhibitory concentration (MIC), mutation frequency (3× MIC), a PK/PD study using the hollow fiber system model of MKC (HFS-MKC) using the reference ATCC#12478 strain, and Monte Carlo simulation experiments for clinical dose selection and susceptibility breakpoint. We also performed a literature search to generate ethambutol MIC distribution for MKC. First, nine studies were identified with MIC of 587 isolates, and MIC<sub>50</sub> and MIC<sub>90</sub> identified as 4 and 16 mg/L, respectively. Second, the ethambutol MIC of the ATCC strain was 8 mg/L, and the mutation frequency was 4.23 × 10<sup>-2</sup> CFU/mL. Third, in the HFS-MKC, ethambutol failed to kill <i>M. kansasii</i> below stasis (<i>B</i><sub>0</sub>), and resistance emerged rapidly. The target exposure was an AUC<sub>0-24</sub>/MIC of 5.47 (95% confidence interval: 1.17-9.77). Fourth, Monte Carlo experiments of 10,000 virtual subjects identified doses of 1,200 and 3,000 mg to achieve or exceed target exposure in 18.21% and 58.57% of patients; and PK/PD MIC susceptibility breakpoints were determined as 2 and 4 mg/L, respectively. Doses >1,200 mg/day may have a higher likelihood of ocular toxicity. The risk of toxicity versus no microbial kill benefit in HFS-MKC suggests the need for better drugs compared to ethambutol in the treatment of MKC lung disease.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0144725"},"PeriodicalIF":4.5,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145793098","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Inhaled liposomal ciprofloxacin protects against lethal tularemia in the common marmoset. 吸入环丙沙星脂质体对普通狨猴的致命性土拉菌病有保护作用。
IF 4.5 2区 医学 Q2 MICROBIOLOGY Pub Date : 2025-12-19 DOI: 10.1128/aac.01232-25
Rachel E Ireland, Alejandro Nunez, Wendy Butcher, Carwyn Davies, James D Blanchard, Francis Dayton, Igor Gonda, Sarah V Harding, Michelle Nelson

Francisella tularensis is a gram-negative, intracellular bacterium that causes the disease tularemia. Tularemia is prevalent in North America, Europe, and Asia and is typically treated with injected and orally administered antibiotics, including streptomycin, gentamicin, doxycycline, and ciprofloxacin, administered for 10 to 21 days. New therapeutic options are required to reduce the potential of a relapse of disease. Inhaled liposomal-encapsulated ciprofloxacin has demonstrated protection in a murine model of tularemia. The efficacy was further assessed in a nonhuman primate model of tularemia. Mixed-sex common marmosets were challenged with F. tularensis by the inhalational route, and the efficacy of ciprofloxacin delivered by either the inhalational (Apulmiq liposomal formulation) or oral route was compared. Antibiotics were initiated either at 24 h post-challenge (post-exposure prophylaxis) or at the onset of fever (treatment) and continued for 7 days. All control (untreated) animals succumbed to infection by 8 days post-challenge. All animals that received antibiotics, by either route, survived the duration of the study, with bacterial clearance in all but one animal that received inhalational ciprofloxacin. Antibiotic treatment also reduced the physiological and immunological responses observed when compared to animals that received no antibiotics. Histological changes in the lungs were less frequent, although mild, resolving lesions were present in animals treated with ciprofloxacin delivered at the onset of fever by either route.

土拉菌弗朗西斯菌是一种革兰氏阴性的细胞内细菌,可引起土拉菌病。土拉菌病在北美、欧洲和亚洲很流行,通常用注射和口服抗生素治疗,包括链霉素、庆大霉素、强力霉素和环丙沙星,疗程10至21天。需要新的治疗方案来减少疾病复发的可能性。吸入脂质体包封环丙沙星在兔吸虫病小鼠模型中显示出保护作用。在兔热病的非人灵长类动物模型中进一步评估了其疗效。采用吸入法对混合性别普通狨猴进行土拉菌感染,比较吸入法(阿普米克脂质体制剂)和口服法给药环丙沙星的效果。在感染后24小时(暴露后预防)或开始发烧(治疗)时开始使用抗生素,并持续7天。所有对照(未治疗)动物在攻毒后8天死于感染。所有接受抗生素治疗的动物,无论是哪种方式,在研究期间都存活了下来,除了一只接受吸入环丙沙星治疗的动物外,其他动物的细菌都被清除了。与未接受抗生素治疗的动物相比,抗生素治疗还减少了观察到的生理和免疫反应。在发烧开始时通过两种途径给予环丙沙星治疗的动物,肺部组织学改变较少,尽管有轻微的消退性病变。
{"title":"Inhaled liposomal ciprofloxacin protects against lethal tularemia in the common marmoset.","authors":"Rachel E Ireland, Alejandro Nunez, Wendy Butcher, Carwyn Davies, James D Blanchard, Francis Dayton, Igor Gonda, Sarah V Harding, Michelle Nelson","doi":"10.1128/aac.01232-25","DOIUrl":"https://doi.org/10.1128/aac.01232-25","url":null,"abstract":"<p><p><i>Francisella tularensis</i> is a gram-negative, intracellular bacterium that causes the disease tularemia. Tularemia is prevalent in North America, Europe, and Asia and is typically treated with injected and orally administered antibiotics, including streptomycin, gentamicin, doxycycline, and ciprofloxacin, administered for 10 to 21 days. New therapeutic options are required to reduce the potential of a relapse of disease. Inhaled liposomal-encapsulated ciprofloxacin has demonstrated protection in a murine model of tularemia. The efficacy was further assessed in a nonhuman primate model of tularemia. Mixed-sex common marmosets were challenged with <i>F. tularensis</i> by the inhalational route, and the efficacy of ciprofloxacin delivered by either the inhalational (Apulmiq liposomal formulation) or oral route was compared. Antibiotics were initiated either at 24 h post-challenge (post-exposure prophylaxis) or at the onset of fever (treatment) and continued for 7 days. All control (untreated) animals succumbed to infection by 8 days post-challenge. All animals that received antibiotics, by either route, survived the duration of the study, with bacterial clearance in all but one animal that received inhalational ciprofloxacin. Antibiotic treatment also reduced the physiological and immunological responses observed when compared to animals that received no antibiotics. Histological changes in the lungs were less frequent, although mild, resolving lesions were present in animals treated with ciprofloxacin delivered at the onset of fever by either route.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0123225"},"PeriodicalIF":4.5,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145793092","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Evaluation of clofazimine-bedaquiline combination as a candidate regimen for macrolide-resistant Mycobacterium avium complex infection. 氯法齐明-贝达喹啉联合治疗大环内酯耐药鸟分枝杆菌复合体感染的候选方案评价。
IF 4.5 2区 医学 Q2 MICROBIOLOGY Pub Date : 2025-12-19 DOI: 10.1128/aac.01511-25
Jiyun Park, Sangwon Choi, Yae Rin Jeon, Lee-Han Kim, Ju Mi Lee, Sung Jae Shin

The Mycobacterium avium complex (MAC) is the primary cause of pulmonary disease (PD) among nontuberculous mycobacteria, presenting a significant treatment challenge on a global scale. A long-term (≥12 months) three-drug regimen, typically including a macrolide, such as clarithromycin (CLR) or azithromycin, along with rifampicin and ethambutol, is recommended. However, many patients fail to respond adequately to therapy, and some eventually develop macrolide resistance, making the disease even more difficult to treat. This highlights the urgent need for improved therapeutic strategies. Here, we investigated the efficacy of clofazimine (CFZ) and bedaquiline (BDQ), both repurposed from multidrug-resistant tuberculosis therapy, against macrolide-resistant MAC. In macrophage infection assays, both CFZ and BDQ showed significant intracellular inhibitory activity against macrolide-resistant clinical isolates, with CFZ generally exhibiting stronger effects. In a chronic murine model of MAC-caused progressive PD, substitution of CLR with CFZ and BDQ in the treatment regimen led to marked reductions in bacterial loads in both lung and spleen compared with the standard regimen, achieving up to 0.86 log₁₀ CFU reduction in lung and 2.17 log₁₀ CFU in spleen tissues. These findings demonstrate that CFZ and BDQ retain potent activity against macrolide-resistant MAC and highlight their potential as promising components of alternative treatment regimens.

鸟分枝杆菌复合体(MAC)是非结核分枝杆菌中肺部疾病(PD)的主要原因,在全球范围内提出了重大的治疗挑战。推荐长期(≥12个月)的三药治疗方案,通常包括大环内酯类药物,如克拉霉素(CLR)或阿奇霉素,以及利福平和乙胺丁醇。然而,许多患者对治疗没有充分的反应,一些患者最终产生大环内酯类药物耐药性,使疾病更加难以治疗。这凸显了改进治疗策略的迫切需要。在这里,我们研究了氯法齐明(CFZ)和贝达喹啉(BDQ)对大环内酯耐药MAC的疗效,这两种药物都是从耐多药结核病治疗中重新利用的。在巨噬细胞感染试验中,CFZ和BDQ对大环内酯耐药临床分离株都表现出显著的细胞内抑制活性,CFZ通常表现出更强的作用。在mac引起的进行性PD的慢性小鼠模型中,与标准方案相比,治疗方案中用CFZ和BDQ替代CLR导致肺和脾脏细菌负荷显著减少,肺组织减少0.86 log₁₀CFU,脾脏组织减少2.17 log₁₀CFU。这些发现表明,CFZ和BDQ对大环内酯耐药的MAC具有有效的活性,并突出了它们作为替代治疗方案的有希望的组成部分的潜力。
{"title":"Evaluation of clofazimine-bedaquiline combination as a candidate regimen for macrolide-resistant <i>Mycobacterium avium</i> complex infection.","authors":"Jiyun Park, Sangwon Choi, Yae Rin Jeon, Lee-Han Kim, Ju Mi Lee, Sung Jae Shin","doi":"10.1128/aac.01511-25","DOIUrl":"https://doi.org/10.1128/aac.01511-25","url":null,"abstract":"<p><p>The <i>Mycobacterium avium</i> complex (MAC) is the primary cause of pulmonary disease (PD) among nontuberculous mycobacteria, presenting a significant treatment challenge on a global scale. A long-term (≥12 months) three-drug regimen, typically including a macrolide, such as clarithromycin (CLR) or azithromycin, along with rifampicin and ethambutol, is recommended. However, many patients fail to respond adequately to therapy, and some eventually develop macrolide resistance, making the disease even more difficult to treat. This highlights the urgent need for improved therapeutic strategies. Here, we investigated the efficacy of clofazimine (CFZ) and bedaquiline (BDQ), both repurposed from multidrug-resistant tuberculosis therapy, against macrolide-resistant MAC. In macrophage infection assays, both CFZ and BDQ showed significant intracellular inhibitory activity against macrolide-resistant clinical isolates, with CFZ generally exhibiting stronger effects. In a chronic murine model of MAC-caused progressive PD, substitution of CLR with CFZ and BDQ in the treatment regimen led to marked reductions in bacterial loads in both lung and spleen compared with the standard regimen, achieving up to 0.86 log₁₀ CFU reduction in lung and 2.17 log₁₀ CFU in spleen tissues. These findings demonstrate that CFZ and BDQ retain potent activity against macrolide-resistant MAC and highlight their potential as promising components of alternative treatment regimens.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0151125"},"PeriodicalIF":4.5,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145793006","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Monthly dalbavancin dosing for suppressive therapy: a pharmacokinetic estimation analysis and case series. 抑制治疗每月剂量达巴文星:药代动力学估计分析和病例系列。
IF 4.5 2区 医学 Q2 MICROBIOLOGY Pub Date : 2025-12-19 DOI: 10.1128/aac.01092-25
Wesley D Kufel, Kristen Tudahl, Paul Hutson, Rahul Mahapatra, Warren Rose, Cecilia Volk

Retained hardware/prosthetic infections frequently require antimicrobial suppression therapy. Treatment options are often limited by resistance, allergies, and dosing frequencies. Dalbavancin (DAL) is a potentially attractive option for suppression of gram-positive infections given its potential for infrequent dosing. However, optimal DAL suppression dosing is unknown. An in silico pharmacokinetic/pharmacodynamic simulation was performed to assess the predicted dalbavancin concentration resulting from suppressive regimens. Serum levels were deemed adequate if the fAUC24/MIC was above the PK target of 27.1. Patients at a U.S. medical center receiving DAL as suppressive therapy were reviewed. PK simulation of DAL dosed 1,500 mg monthly resulted in free serum concentrations above the PK target. Because many clinicians opt to initiate these regimens with two doses given 1 week apart, the next modeled regimen included this load, before initiating 1,500 mg monthly. This initial load did not significantly alter total drug exposure. The final simulated regimen was 1,000 mg monthly. With this simulation, the lower 95% CI fAUC24/MIC fell just below the PK target for an isolate at the breakpoint. Nine patients who received dalbavancin 1,500 mg monthly as suppressive therapy were reviewed. All had retained hardware and received DAL for a median 591 days, with 7 patients still receiving treatment and no reported suppressive therapy failure. Monthly 1500 mg dalbavancin dosing for suppressive therapy is supported by this case series and PK simulation data. An initial weekly loading dose appears unnecessary. Reducing the monthly dose to 1000 mg may also be appropriate for certain patients, though clinical data is needed to support this practice.

保留的硬件/假体感染经常需要抗菌抑制治疗。治疗方案通常受到耐药性、过敏和给药频率的限制。Dalbavancin (DAL)是抑制革兰氏阳性感染的潜在有吸引力的选择,因为它可能不经常给药。然而,最佳的DAL抑制剂量是未知的。进行了计算机药代动力学/药效学模拟,以评估抑制方案导致的dalbavancin预测浓度。如果fAUC24/MIC高于PK目标27.1,则认为血清水平足够。在美国医疗中心接受DAL作为抑制治疗的患者进行了回顾。每月给药1500 mg的DAL进行PK模拟,导致游离血清浓度高于PK目标。因为许多临床医生选择在开始这些方案时,两次剂量间隔一周,下一个模拟方案包括这个负荷,然后开始每月1500毫克。这一初始负荷并没有显著改变总药物暴露。最终模拟方案为每月1000毫克。通过这种模拟,在断点处,较低的95% CI fAUC24/MIC刚好低于隔离物的PK目标。本文回顾了9例每月接受达巴文星1500mg抑制治疗的患者。所有患者均保留了硬体,接受DAL治疗的中位时间为591天,其中7名患者仍在接受治疗,没有报告抑制治疗失败。该病例系列和PK模拟数据支持每月1500mg达巴万星剂量用于抑制治疗。最初的每周负荷剂量似乎没有必要。对于某些患者,将月剂量减少至1000mg也可能是合适的,尽管需要临床数据来支持这种做法。
{"title":"Monthly dalbavancin dosing for suppressive therapy: a pharmacokinetic estimation analysis and case series.","authors":"Wesley D Kufel, Kristen Tudahl, Paul Hutson, Rahul Mahapatra, Warren Rose, Cecilia Volk","doi":"10.1128/aac.01092-25","DOIUrl":"https://doi.org/10.1128/aac.01092-25","url":null,"abstract":"<p><p>Retained hardware/prosthetic infections frequently require antimicrobial suppression therapy. Treatment options are often limited by resistance, allergies, and dosing frequencies. Dalbavancin (DAL) is a potentially attractive option for suppression of gram-positive infections given its potential for infrequent dosing. However, optimal DAL suppression dosing is unknown. An <i>in silico</i> pharmacokinetic/pharmacodynamic simulation was performed to assess the predicted dalbavancin concentration resulting from suppressive regimens. Serum levels were deemed adequate if the <i>f</i>AUC<sub>24</sub>/MIC was above the PK target of 27.1. Patients at a U.S. medical center receiving DAL as suppressive therapy were reviewed. PK simulation of DAL dosed 1,500 mg monthly resulted in free serum concentrations above the PK target. Because many clinicians opt to initiate these regimens with two doses given 1 week apart, the next modeled regimen included this load, before initiating 1,500 mg monthly. This initial load did not significantly alter total drug exposure. The final simulated regimen was 1,000 mg monthly. With this simulation, the lower 95% CI <i>f</i>AUC<sub>24</sub>/MIC fell just below the PK target for an isolate at the breakpoint. Nine patients who received dalbavancin 1,500 mg monthly as suppressive therapy were reviewed. All had retained hardware and received DAL for a median 591 days, with 7 patients still receiving treatment and no reported suppressive therapy failure. Monthly 1500 mg dalbavancin dosing for suppressive therapy is supported by this case series and PK simulation data. An initial weekly loading dose appears unnecessary. Reducing the monthly dose to 1000 mg may also be appropriate for certain patients, though clinical data is needed to support this practice.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0109225"},"PeriodicalIF":4.5,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145793086","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Identification and characterization of a novel inhibitor of influenza A virus that acts by blocking nucleoprotein oligomerization. 通过阻断核蛋白寡聚化作用的新型甲型流感病毒抑制剂的鉴定和表征。
IF 4.5 2区 医学 Q2 MICROBIOLOGY Pub Date : 2025-12-19 DOI: 10.1128/aac.01149-25
Vincent H J Leonard, Dianna B Vidales, Benjamin R Taft, Matthew J Hesse, Patrick S Lee, Mulugeta Mamo, Dirksen E Bussiere, Karen C Wolff, Kelli L Kuhen, Laura Wedel, Ellena Growcott, Colin Osborne, Cassio P Octaviani, Pinghan Huang, Chien-Te Kent Tseng, Johanna R Abend, Kelly A Wong, Weidong Zhong, David C Tully, Don Ganem

Influenza A virus (IAV) causes annual epidemics and sporadic pandemics of acute respiratory infections resulting in significant morbidity and mortality. Although approved influenza antivirals (e.g., oseltamivir and baloxavir) exist, concerns persist about the potential for emergence of drug-resistant variants, highlighting the continuing need for new antiviral therapies. Here, we describe the development of an orally bioavailable, direct-acting antiviral (VNT-101) with a novel mechanism of action: disrupting homo-oligomerization of the influenza nucleoprotein (NP) and thereby inhibiting viral RNA synthesis. Selection of drug-resistant mutants revealed amino acid substitutions mapping to the oligomerization domain of NP, and X-ray crystallography co-structure determination of VNT-101 complexed with recombinant NP confirmed VNT-101 binding in the oligomerization pocket. Biochemical experiments using size exclusion chromatography confirmed disruption of oligomerization when this chemotype is added to preparations of recombinant NP in vitro. VNT-101 has potent and specific activity against the currently circulating IAV subtypes H1N1 and H3N2, with mean EC50 values ranging from 2 to 18 nM, and displays strong efficacy in a murine model of lethal influenza infection when administered either prophylactically or therapeutically. Importantly, VNT-101 remains active against influenza variants that are resistant to either oseltamivir or baloxavir and also has potent activity against highly pathogenic avian H5N1 and H7N9 isolates that have transmitted to humans and represent strains of potential pandemic concern. These studies support the continued development of VNT-101 to augment our therapeutic arsenal against both seasonal and pandemic influenza.

甲型流感病毒(IAV)引起急性呼吸道感染的年度流行和散发大流行,造成严重的发病率和死亡率。虽然已有批准的流感抗病毒药物(如奥司他韦和巴洛昔韦),但人们仍然担心出现耐药变异的可能性,这突出表明需要继续开发新的抗病毒疗法。在这里,我们描述了一种口服生物可用的直接作用抗病毒药物(VNT-101)的发展,它具有一种新的作用机制:破坏流感核蛋白(NP)的同质寡聚,从而抑制病毒RNA的合成。对耐药突变体的筛选发现,氨基酸取代映射到NP的寡聚化结构域,x射线晶体学检测发现,VNT-101与重组NP配合后,证实了VNT-101在寡聚化袋中结合。采用粒径排除色谱法进行的生化实验证实,将这种化学型添加到体外重组NP的制备中会破坏寡聚化。VNT-101对当前流行的IAV亚型H1N1和H3N2具有强效和特异性活性,平均EC50值在2至18 nM之间,并且在小鼠致死性流感感染模型中,无论是预防性给药还是治疗性给药,都显示出很强的疗效。重要的是,VNT-101对奥司他韦或巴洛昔韦耐药的流感变种仍有活性,对已传播给人类的高致病性H5N1和H7N9禽流感分离株也有强效活性,这两种病毒可能引起大流行。这些研究支持继续开发VNT-101,以增强我们针对季节性和大流行性流感的治疗武器库。
{"title":"Identification and characterization of a novel inhibitor of influenza A virus that acts by blocking nucleoprotein oligomerization.","authors":"Vincent H J Leonard, Dianna B Vidales, Benjamin R Taft, Matthew J Hesse, Patrick S Lee, Mulugeta Mamo, Dirksen E Bussiere, Karen C Wolff, Kelli L Kuhen, Laura Wedel, Ellena Growcott, Colin Osborne, Cassio P Octaviani, Pinghan Huang, Chien-Te Kent Tseng, Johanna R Abend, Kelly A Wong, Weidong Zhong, David C Tully, Don Ganem","doi":"10.1128/aac.01149-25","DOIUrl":"https://doi.org/10.1128/aac.01149-25","url":null,"abstract":"<p><p>Influenza A virus (IAV) causes annual epidemics and sporadic pandemics of acute respiratory infections resulting in significant morbidity and mortality. Although approved influenza antivirals (e.g., oseltamivir and baloxavir) exist, concerns persist about the potential for emergence of drug-resistant variants, highlighting the continuing need for new antiviral therapies. Here, we describe the development of an orally bioavailable, direct-acting antiviral (VNT-101) with a novel mechanism of action: disrupting homo-oligomerization of the influenza nucleoprotein (NP) and thereby inhibiting viral RNA synthesis. Selection of drug-resistant mutants revealed amino acid substitutions mapping to the oligomerization domain of NP, and X-ray crystallography co-structure determination of VNT-101 complexed with recombinant NP confirmed VNT-101 binding in the oligomerization pocket. Biochemical experiments using size exclusion chromatography confirmed disruption of oligomerization when this chemotype is added to preparations of recombinant NP <i>in vitro</i>. VNT-101 has potent and specific activity against the currently circulating IAV subtypes H1N1 and H3N2, with mean EC<sub>50</sub> values ranging from 2 to 18 nM, and displays strong efficacy in a murine model of lethal influenza infection when administered either prophylactically or therapeutically. Importantly, VNT-101 remains active against influenza variants that are resistant to either oseltamivir or baloxavir and also has potent activity against highly pathogenic avian H5N1 and H7N9 isolates that have transmitted to humans and represent strains of potential pandemic concern. These studies support the continued development of VNT-101 to augment our therapeutic arsenal against both seasonal and pandemic influenza.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0114925"},"PeriodicalIF":4.5,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145793052","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Isobavachalcone effectively inhibits the growth of Candida albicans. 异巴伐恰尔酮有效抑制白色念珠菌的生长。
IF 4.5 2区 医学 Q2 MICROBIOLOGY Pub Date : 2025-12-19 DOI: 10.1128/aac.00797-25
Ping Xie, Wenting Zhou, Jiazi Luo, Yuanyuan Dai, Shixian Yang, Shufang Li, Yanqiang Huang

Candida albicans (C. albicans) is a commensal, drug-resistant opportunistic pathogen, and the eradication of invasive candidiasis represents a significant clinical challenge. This study investigated the inhibitory effect of isobavachalcone (IBC) on C. albicans growth and elucidated its mechanism. The antifungal activity of IBC was evaluated using minimum inhibitory concentration 90% (MIC90) and minimum fungicidal concentration (MFC), combined with murine vaginitis and oral thrush models to assess in vivo efficacy. An MTT assay was used to assess drug safety. Mechanistic investigations included cell membrane/wall damage assessments, virulence factor inhibition, oxidative stress evaluation, ATP metabolism analysis, protein expression profiling, and target identification (including RT-qPCR, inhibitor intervention experiments, and related methodologies). The antifungal potency of IBC against C. albicans was demonstrated with a MIC90 of 2 µg/mL and an MFC of 8 μg/mL. Against strain S393, IBC exhibited potent efficacy with median effective and effective concentrations of 1.301 µg/mL and 1.449 µg/mL, respectively. In vivo, IBC significantly alleviated vulvovaginal candidiasis and oropharyngeal thrush, outperforming fluconazole in therapeutic efficacy and mucosal protection. Mechanistic studies revealed that IBC prevented fungal invasion by inhibiting C. albicans adhesion and colonization on mucosal surfaces, mitigated inflammation through suppression of proinflammatory cytokine release, and downregulated the expression of ADE13, TPI1, and ADK1 genes, with ADK1 demonstrating the most significant suppression. Furthermore, IBC specifically bound to ADK1, inhibiting ATP synthesis and disrupting cellular energy metabolism in C. albicans. IBC represents a promising antifungal drug that acts by downregulating the ADE13, TPI1, and ADK1 genes. Its downregulation of ADK1 leads to impaired ATP synthesis, induced DNA damage, and suppressed fungal proliferation.

白色念珠菌(C. albicans)是一种共生的、耐药的机会性病原体,根除侵袭性念珠菌病是一项重大的临床挑战。本研究研究了异巴伐恰尔酮(IBC)对白色念珠菌生长的抑制作用,并阐明了其作用机制。采用最小抑菌浓度90% (MIC90)和最小杀真菌浓度(MFC)评价IBC的抗真菌活性,并结合小鼠阴道炎和鹅口疮模型评价IBC的体内药效。采用MTT法评价药物安全性。机制研究包括细胞膜/细胞壁损伤评估、毒力因子抑制、氧化应激评估、ATP代谢分析、蛋白表达谱和靶标鉴定(包括RT-qPCR、抑制剂干预实验和相关方法)。IBC对白色念珠菌的MIC90为2 μg/mL, MFC为8 μg/mL。IBC对菌株S393的中位有效浓度为1.301µg/mL,有效浓度为1.449µg/mL。在体内,IBC显著缓解外阴阴道念珠菌病和口咽鹅口疮,在治疗效果和粘膜保护方面优于氟康唑。机制研究表明,IBC通过抑制白色念珠菌在粘膜表面的粘附和定植来阻止真菌的侵袭,通过抑制促炎细胞因子的释放来减轻炎症,并下调ADE13、TPI1和ADK1基因的表达,其中ADK1的抑制作用最为显著。此外,IBC特异性结合ADK1,抑制ATP合成并破坏白色念珠菌的细胞能量代谢。IBC是一种很有前景的抗真菌药物,它通过下调ADE13、TPI1和ADK1基因起作用。其下调ADK1导致ATP合成受损,诱导DNA损伤,抑制真菌增殖。
{"title":"Isobavachalcone effectively inhibits the growth of <i>Candida albicans</i>.","authors":"Ping Xie, Wenting Zhou, Jiazi Luo, Yuanyuan Dai, Shixian Yang, Shufang Li, Yanqiang Huang","doi":"10.1128/aac.00797-25","DOIUrl":"https://doi.org/10.1128/aac.00797-25","url":null,"abstract":"<p><p><i>Candida albicans</i> (<i>C. albicans</i>) is a commensal, drug-resistant opportunistic pathogen, and the eradication of invasive candidiasis represents a significant clinical challenge. This study investigated the inhibitory effect of isobavachalcone (IBC) on <i>C. albicans</i> growth and elucidated its mechanism. The antifungal activity of IBC was evaluated using minimum inhibitory concentration 90% (MIC<sub>90</sub>) and minimum fungicidal concentration (MFC), combined with murine vaginitis and oral thrush models to assess <i>in vivo</i> efficacy. An MTT assay was used to assess drug safety. Mechanistic investigations included cell membrane/wall damage assessments, virulence factor inhibition, oxidative stress evaluation, ATP metabolism analysis, protein expression profiling, and target identification (including RT-qPCR, inhibitor intervention experiments, and related methodologies). The antifungal potency of IBC against <i>C. albicans</i> was demonstrated with a MIC<sub>90</sub> of 2 µg/mL and an MFC of 8 μg/mL. Against strain S393, IBC exhibited potent efficacy with median effective and effective concentrations of 1.301 µg/mL and 1.449 µg/mL, respectively. <i>In vivo</i>, IBC significantly alleviated vulvovaginal candidiasis and oropharyngeal thrush, outperforming fluconazole in therapeutic efficacy and mucosal protection. Mechanistic studies revealed that IBC prevented fungal invasion by inhibiting <i>C. albicans</i> adhesion and colonization on mucosal surfaces, mitigated inflammation through suppression of proinflammatory cytokine release, and downregulated the expression of <i>ADE13</i>, <i>TPI1</i>, and <i>ADK1</i> genes, with <i>ADK1</i> demonstrating the most significant suppression. Furthermore, IBC specifically bound to ADK1, inhibiting ATP synthesis and disrupting cellular energy metabolism in <i>C. albicans</i>. IBC represents a promising antifungal drug that acts by downregulating the <i>ADE13</i>, <i>TPI1</i>, and <i>ADK1</i> genes. Its downregulation of ADK1 leads to impaired ATP synthesis, induced DNA damage, and suppressed fungal proliferation.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0079725"},"PeriodicalIF":4.5,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145793059","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Essential role of MHC II in the antitubercular efficacy of pyrazinamide. MHCⅱ在吡嗪酰胺抗结核疗效中的重要作用。
IF 4.5 2区 医学 Q2 MICROBIOLOGY Pub Date : 2025-12-19 DOI: 10.1128/aac.01264-25
Elise A Lamont, Shannon L Kordus, Michael D Howe, Ziyi Jia, Nathan Schacht, Muzafar Ahmad Rather, Gebremichal Gebretsadik, Anthony D Baughn

Antibacterial drug mechanisms have traditionally been examined through a drug-pathogen lens, with limited attention to host influences on drug activity. However, growing evidence suggests that the host environment is crucial for antibacterial efficacy. Pyrazinamide (PZA), a key component of modern tuberculosis therapy, exemplifies this complexity, exhibiting potent in vivo activity despite its inability to reduce Mycobacterium tuberculosis viability in standard in vitro culture. Here, using macrophage and murine infection models, we identify a critical role for CD4+ T cell-dependent cell-mediated immunity in PZA's antitubercular action. Using MHC class II knockout mice, we demonstrate that CD4 T-cell help is essential for PZA efficacy. While interferon gamma (IFN-γ) is required for PZA-mediated clearance of M. tuberculosis at extrapulmonary sites, bacterial reduction in the lungs occurs, independent of IFN-γ signaling. We show that PZA leverages cell-mediated immunity in part through activation of the oxidative burst. Our findings underscore the need to incorporate host factors into antibacterial drug evaluation and highlight potential avenues for host-directed therapies and adjunctive antibiotics in first- and second-line tuberculosis treatment.

传统上,抗菌药物的机制是通过药物-病原体透镜来研究的,对宿主对药物活性的影响关注有限。然而,越来越多的证据表明,宿主环境对抗菌效果至关重要。吡嗪酰胺(PZA)是现代结核病治疗的关键成分,体现了这种复杂性,尽管在标准体外培养中无法降低结核分枝杆菌的活力,但它在体内表现出强大的活性。在这里,利用巨噬细胞和小鼠感染模型,我们确定了CD4+ T细胞依赖的细胞介导免疫在PZA抗结核作用中的关键作用。使用MHC II类敲除小鼠,我们证明CD4 t细胞的帮助对PZA的疗效至关重要。虽然pza介导的肺外部位结核分枝杆菌的清除需要干扰素γ (IFN-γ),但肺部细菌的减少是独立于IFN-γ信号的。我们发现PZA部分通过激活氧化爆发来利用细胞介导的免疫。我们的研究结果强调了将宿主因素纳入抗菌药物评估的必要性,并强调了宿主导向疗法和辅助抗生素在一线和二线结核病治疗中的潜在途径。
{"title":"Essential role of MHC II in the antitubercular efficacy of pyrazinamide.","authors":"Elise A Lamont, Shannon L Kordus, Michael D Howe, Ziyi Jia, Nathan Schacht, Muzafar Ahmad Rather, Gebremichal Gebretsadik, Anthony D Baughn","doi":"10.1128/aac.01264-25","DOIUrl":"10.1128/aac.01264-25","url":null,"abstract":"<p><p>Antibacterial drug mechanisms have traditionally been examined through a drug-pathogen lens, with limited attention to host influences on drug activity. However, growing evidence suggests that the host environment is crucial for antibacterial efficacy. Pyrazinamide (PZA), a key component of modern tuberculosis therapy, exemplifies this complexity, exhibiting potent <i>in vivo</i> activity despite its inability to reduce <i>Mycobacterium tuberculosis</i> viability in standard <i>in vitro</i> culture. Here, using macrophage and murine infection models, we identify a critical role for CD4<sup>+</sup> T cell-dependent cell-mediated immunity in PZA's antitubercular action. Using MHC class II knockout mice, we demonstrate that CD4 T-cell help is essential for PZA efficacy. While interferon gamma (IFN-γ) is required for PZA-mediated clearance of <i>M. tuberculosis</i> at extrapulmonary sites, bacterial reduction in the lungs occurs, independent of IFN-γ signaling. We show that PZA leverages cell-mediated immunity in part through activation of the oxidative burst. Our findings underscore the need to incorporate host factors into antibacterial drug evaluation and highlight potential avenues for host-directed therapies and adjunctive antibiotics in first- and second-line tuberculosis treatment.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0126425"},"PeriodicalIF":4.5,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145793100","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Antimicrobial Agents and Chemotherapy
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
×
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
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
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