Pub Date : 2025-12-10DOI: 10.1016/j.ijantimicag.2025.107691
Abdullah T Aslan, Patrick N A Harris, David L Paterson
{"title":"In reply to the Letter to Editor Regarding \"Comparison of ceftazidime-avibactam with other appropriate antimicrobial therapy for the treatment of OXA-48- or KPC-producing Enterobacterales infections in Turkiye: A multi-centre retrospective matched-cohort study\".","authors":"Abdullah T Aslan, Patrick N A Harris, David L Paterson","doi":"10.1016/j.ijantimicag.2025.107691","DOIUrl":"https://doi.org/10.1016/j.ijantimicag.2025.107691","url":null,"abstract":"","PeriodicalId":13818,"journal":{"name":"International Journal of Antimicrobial Agents","volume":" ","pages":"107691"},"PeriodicalIF":4.6,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145742497","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}
Pub Date : 2025-12-10DOI: 10.1016/j.ijantimicag.2025.107692
Alba Pau-Parra, Sònia Luque, Laura Doménech, María Martínez-Pla, Xavier Nuvials, Manuel Sosa Garay, Ibai Los-Arcos, Aldair Conto, Fernando Lasteros, Elisabet Gallart, Maria Queralt Gorgas Torner, Ricard Ferrer, Jordi Riera
{"title":"Aztreonam-avibactam pharmacokinetics during extracorporeal membrane oxygenation support: first insights from a case report.","authors":"Alba Pau-Parra, Sònia Luque, Laura Doménech, María Martínez-Pla, Xavier Nuvials, Manuel Sosa Garay, Ibai Los-Arcos, Aldair Conto, Fernando Lasteros, Elisabet Gallart, Maria Queralt Gorgas Torner, Ricard Ferrer, Jordi Riera","doi":"10.1016/j.ijantimicag.2025.107692","DOIUrl":"https://doi.org/10.1016/j.ijantimicag.2025.107692","url":null,"abstract":"","PeriodicalId":13818,"journal":{"name":"International Journal of Antimicrobial Agents","volume":" ","pages":"107692"},"PeriodicalIF":4.6,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145742533","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}
Pub Date : 2025-12-10DOI: 10.1016/j.ijantimicag.2025.107690
Jie Xu, Yuan Li, Ying Wang, Fang Liu, Jinzhao Long, Jingyuan Zhu, Yuefei Jin, Shuaiyin Chen, Guangcai Duan, Haiyan Yang
Introduction: The evolution of antimicrobial resistance (AMR) in Staphylococcus aureus (S. aureus) involves complex genotype-phenotype interactions of multiple genetic determinants acting independently.
Objectives: In this study, we aimed to accurately predict AMR from genomic sequences and uncover the complex genetic interactions underlying its mechanisms, focusing on interpretability and mechanistic insights.
Methods: We propose a framework that elucidates antibiotic resistance by linking the genomic context with underlying gene interactions, combining a reference-agnostic gene-context 22-mer (gkmer) representation, a two-step random forest pipeline (RF1 screening/mapping; RF2 gene-level modeling), co-information-based synergy networks, and protein-structure mapping.
Results: Using genomic sequences from 4,569 S. aureus strains, we successfully predicted resistance to 12 antibiotics and examined the genetic interaction networks for penicillin, ciprofloxacin, and erythromycin in detail. On the held-out 20% test set, RF2 achieved an area under the receiver operating characteristic curve (AUC) of 0.83-1.00 (median 0.90) and an F1 score of 0.58-0.99 (median 0.76); comparable performance was observed for RF1. External validation on an independent dataset including 1,011 S. aureus isolates revealed strong generalization for ciprofloxacin, erythromycin, and penicillin (AUC > 0.95; F1 > 0.97), while highlighting reduced performance for clindamycin and tetracycline, and failure for gentamicin and trimethoprim, thereby delineating the scope and limits of the model and its applicability. Network analysis revealed distinct structural patterns of gene cooperation, highlighting key hubs and community structures consistent with known resistance pathways.
Conclusion: Our findings indicate that antibiotic resistance in S. aureus involves distinct genetic network architectures depending on the antibiotic, highlighting the diverse regulatory pathways bacteria employ to acquire resistance. This discovery-oriented framework prioritizes interpretable determinants and generates testable mechanistic hypotheses, with prospective standardized evaluations aimed at assessing its translatability to clinical settings.
简介:金黄色葡萄球菌(S. aureus)抗微生物药物耐药性(AMR)的进化涉及多个独立作用的遗传决定因素的复杂基因型-表型相互作用。目的:在本研究中,我们旨在从基因组序列中准确预测AMR,揭示其复杂的遗传相互作用机制,重点关注其可解释性和机制见解。方法:我们提出了一个框架,通过将基因组背景与潜在的基因相互作用联系起来,结合参考不可知的基因背景22-mer (gkmer)表示,两步随机森林管道(RF1筛选/定位;RF2基因水平建模),基于共同信息的协同网络和蛋白质结构定位来阐明抗生素耐药性。结果:利用4569株金黄色葡萄球菌的基因组序列,我们成功预测了12种抗生素的耐药性,并详细检测了青霉素、环丙沙星和红霉素的遗传相互作用网络。在hold out 20%的测试集上,RF2的受试者工作特征曲线下面积(AUC)为0.83-1.00(中位数0.90),F1得分为0.58-0.99(中位数0.76);RF1也有类似的表现。在包括1011株金黄色葡萄球菌分离物的独立数据集上进行的外部验证显示,环丙沙星、红霉素和青霉素具有很强的泛化性(AUC > 0.95; F1 > 0.97),同时强调克林霉素和四环素的性能降低,庆大霉素和甲氧苄啶的性能失败,从而描绘了模型的范围和局限性及其适用性。网络分析揭示了不同的基因合作结构模式,突出了与已知抗性途径一致的关键枢纽和群落结构。结论:我们的研究结果表明,金黄色葡萄球菌的抗生素耐药性涉及不同的遗传网络结构,这取决于抗生素,突出了细菌获得耐药性的不同调控途径。这个以发现为导向的框架优先考虑可解释的决定因素,并产生可测试的机制假设,具有前瞻性的标准化评估,旨在评估其在临床环境中的可翻译性。
{"title":"Machine-Learning and Gene-Synergy Networks Reveal Interpretable Drivers of Antibiotic Resistance in Staphylococcus aureus.","authors":"Jie Xu, Yuan Li, Ying Wang, Fang Liu, Jinzhao Long, Jingyuan Zhu, Yuefei Jin, Shuaiyin Chen, Guangcai Duan, Haiyan Yang","doi":"10.1016/j.ijantimicag.2025.107690","DOIUrl":"https://doi.org/10.1016/j.ijantimicag.2025.107690","url":null,"abstract":"<p><strong>Introduction: </strong>The evolution of antimicrobial resistance (AMR) in Staphylococcus aureus (S. aureus) involves complex genotype-phenotype interactions of multiple genetic determinants acting independently.</p><p><strong>Objectives: </strong>In this study, we aimed to accurately predict AMR from genomic sequences and uncover the complex genetic interactions underlying its mechanisms, focusing on interpretability and mechanistic insights.</p><p><strong>Methods: </strong>We propose a framework that elucidates antibiotic resistance by linking the genomic context with underlying gene interactions, combining a reference-agnostic gene-context 22-mer (gkmer) representation, a two-step random forest pipeline (RF1 screening/mapping; RF2 gene-level modeling), co-information-based synergy networks, and protein-structure mapping.</p><p><strong>Results: </strong>Using genomic sequences from 4,569 S. aureus strains, we successfully predicted resistance to 12 antibiotics and examined the genetic interaction networks for penicillin, ciprofloxacin, and erythromycin in detail. On the held-out 20% test set, RF2 achieved an area under the receiver operating characteristic curve (AUC) of 0.83-1.00 (median 0.90) and an F1 score of 0.58-0.99 (median 0.76); comparable performance was observed for RF1. External validation on an independent dataset including 1,011 S. aureus isolates revealed strong generalization for ciprofloxacin, erythromycin, and penicillin (AUC > 0.95; F1 > 0.97), while highlighting reduced performance for clindamycin and tetracycline, and failure for gentamicin and trimethoprim, thereby delineating the scope and limits of the model and its applicability. Network analysis revealed distinct structural patterns of gene cooperation, highlighting key hubs and community structures consistent with known resistance pathways.</p><p><strong>Conclusion: </strong>Our findings indicate that antibiotic resistance in S. aureus involves distinct genetic network architectures depending on the antibiotic, highlighting the diverse regulatory pathways bacteria employ to acquire resistance. This discovery-oriented framework prioritizes interpretable determinants and generates testable mechanistic hypotheses, with prospective standardized evaluations aimed at assessing its translatability to clinical settings.</p>","PeriodicalId":13818,"journal":{"name":"International Journal of Antimicrobial Agents","volume":" ","pages":"107690"},"PeriodicalIF":4.6,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145742502","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}
Pub Date : 2025-12-09DOI: 10.1016/j.ijantimicag.2025.107689
Ziyuan Zhao, Leshan Xiu, Junping Peng
The introduction of doxycycline post-exposure prophylaxis (doxy-PEP) as a strategy to reduce the risk of bacterial sexually transmitted infections has raised concerns about the potential for rapid selection of tetracycline resistance and the emergence of ceftriaxone-resistant strains. This study aimed to develop a molecular surveillance method to monitor antimicrobial resistance (AMR) in Neisseria gonorrhoeae (NG) during doxy-PEP implementation. We developed a high-resolution melting (HRM) analysis-based molecular assay, termed HRM-doxyPEP, to enhance surveillance of NG AMR. Validation was conducted with strains of known susceptibility profiles, and further evaluation was performed on clinical samples, comparing HRM results with qPCR, whole-genome sequencing and PCR-sequencing. The HRM-doxyPEP assay demonstrated high analytical specificity and a detection limit of 20 copies/reaction, enabling the detection of low levels of target DNA in clinical samples. Validation with strains of known susceptibility profiles showed 84% consistency with AST results. Clinical sample testing revealed 100% sensitivity and specificity for identifying NG infection, tracking ceftriaxone-resistant FC428-like strains, and detecting tetM associated with tetracycline resistance. The HRM-doxyPEP assay offers an accurate, affordable, and scalable tool for enhanced molecular surveillance of AMR in NG during doxy-PEP implementation.
{"title":"Enhanced Molecular Surveillance for Gonococcal Resistance During Doxycycline Post-Exposure Prophylaxis Implementation.","authors":"Ziyuan Zhao, Leshan Xiu, Junping Peng","doi":"10.1016/j.ijantimicag.2025.107689","DOIUrl":"https://doi.org/10.1016/j.ijantimicag.2025.107689","url":null,"abstract":"<p><p>The introduction of doxycycline post-exposure prophylaxis (doxy-PEP) as a strategy to reduce the risk of bacterial sexually transmitted infections has raised concerns about the potential for rapid selection of tetracycline resistance and the emergence of ceftriaxone-resistant strains. This study aimed to develop a molecular surveillance method to monitor antimicrobial resistance (AMR) in Neisseria gonorrhoeae (NG) during doxy-PEP implementation. We developed a high-resolution melting (HRM) analysis-based molecular assay, termed HRM-doxyPEP, to enhance surveillance of NG AMR. Validation was conducted with strains of known susceptibility profiles, and further evaluation was performed on clinical samples, comparing HRM results with qPCR, whole-genome sequencing and PCR-sequencing. The HRM-doxyPEP assay demonstrated high analytical specificity and a detection limit of 20 copies/reaction, enabling the detection of low levels of target DNA in clinical samples. Validation with strains of known susceptibility profiles showed 84% consistency with AST results. Clinical sample testing revealed 100% sensitivity and specificity for identifying NG infection, tracking ceftriaxone-resistant FC428-like strains, and detecting tetM associated with tetracycline resistance. The HRM-doxyPEP assay offers an accurate, affordable, and scalable tool for enhanced molecular surveillance of AMR in NG during doxy-PEP implementation.</p>","PeriodicalId":13818,"journal":{"name":"International Journal of Antimicrobial Agents","volume":" ","pages":"107689"},"PeriodicalIF":4.6,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145742543","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}
Pub Date : 2025-12-04DOI: 10.1016/j.ijantimicag.2025.107687
Melike Törüyenler Coşkunpınar, Güle Çınar, Duygu Öcal, İsmail Balık
Background: Carbapenemase-producing Klebsiella pneumoniae (CP-Kp) poses a significant global health threat due to its limited treatment options and increasing resistance. Fosfomycin (FOF) has regained interest, particularly in combination therapies. This study aimed to evaluate the in vitro synergy of FOF with meropenem (MEM) and polymyxin in CP-Kp bloodstream infections (BSI) and to investigate clinical outcomes.
Methods: Fifty-two patients with CP-Kp BSI who received FOF-based combination therapy (FOF-C) for ≥72 hours were evaluated retrospectively for clinical outcomes. MICs were determined on stored isolates: FOF by agar dilution (AD) and broth microdilution (BMD), MEM and COL by BMD. In vitro synergy testing of FOF with MEM and COL was performed using the checkerboard (CB) method. Data were analyzed using logistic regression.
Results: Among 50 isolates, the FOF AD MIC50/90 were 64/>128 μg/mL. Positive in vitro interactions (synergistic or additive) were observed in 43.5% of isolates for both FOF+MEM and FOF+COL, with no antagonism. The clinical and microbiological response rates were 61.5% and 80.5%, 14- and 30-day mortality rates were 21.2% and 40.4%, respectively. Patients receiving at least one agent with in vitro positive interaction had higher clinical response, but the difference wasn't significant (73.7% vs. 52.2% p=0.153). In multivariable analysis, increased FOF AD MIC and Pitt bacteremia score (PBS) were associated with clinical nonresponse, while higher COL MIC and PBS predicted 30-day mortality.
Conclusion: FOF AD MIC value may have potential as a predictive marker for FOF-C treatment response in CP-Kp BSI and could aid in guiding combination therapy decisions. Larger, prospective studies using standardized synergy testing methods are needed.
背景:产碳青霉烯酶肺炎克雷伯菌(CP-Kp)由于其治疗选择有限和耐药性增加,对全球健康构成重大威胁。磷霉素(FOF)已重新引起人们的兴趣,特别是在联合治疗中。本研究旨在评估FOF与美罗培南(MEM)和多粘菌素在CP-Kp血流感染(BSI)中的体外协同作用,并探讨临床结果。方法:回顾性评价52例接受fof联合治疗(FOF-C)≥72小时的CP-Kp BSI患者的临床结果。对储存的分离株进行mic测定:琼脂稀释法(AD)和肉汤微量稀释法(BMD)测定FOF, BMD测定MEM和COL。采用棋盘法测定FOF与MEM、COL的体外协同作用。数据采用逻辑回归分析。结果:50株分离菌FOF AD MIC50/90为64/ bb0 ~ 128 μg/mL;43.5%的分离株对FOF+MEM和FOF+COL均有正向的体外相互作用(协同或加性),无拮抗作用。临床和微生物应答率分别为61.5%和80.5%,14天和30天死亡率分别为21.2%和40.4%。接受至少一种药物体外正相互作用的患者临床疗效较高,但差异无统计学意义(73.7% vs. 52.2% p=0.153)。在多变量分析中,FOF AD MIC和Pitt菌血症评分(PBS)升高与临床无反应相关,而较高的COL MIC和PBS预测30天死亡率。结论:FOF- AD - MIC值可能作为预测CP-Kp BSI患者FOF- c治疗反应的潜在指标,并有助于指导联合治疗决策。需要使用标准化协同测试方法进行更大规模的前瞻性研究。
{"title":"Fosfomycin-based combinations against carbapenemase-producing Klebsiella pneumoniae bloodstream infections: Correlating in vitro synergy with clinical outcomes.","authors":"Melike Törüyenler Coşkunpınar, Güle Çınar, Duygu Öcal, İsmail Balık","doi":"10.1016/j.ijantimicag.2025.107687","DOIUrl":"https://doi.org/10.1016/j.ijantimicag.2025.107687","url":null,"abstract":"<p><strong>Background: </strong>Carbapenemase-producing Klebsiella pneumoniae (CP-Kp) poses a significant global health threat due to its limited treatment options and increasing resistance. Fosfomycin (FOF) has regained interest, particularly in combination therapies. This study aimed to evaluate the in vitro synergy of FOF with meropenem (MEM) and polymyxin in CP-Kp bloodstream infections (BSI) and to investigate clinical outcomes.</p><p><strong>Methods: </strong>Fifty-two patients with CP-Kp BSI who received FOF-based combination therapy (FOF-C) for ≥72 hours were evaluated retrospectively for clinical outcomes. MICs were determined on stored isolates: FOF by agar dilution (AD) and broth microdilution (BMD), MEM and COL by BMD. In vitro synergy testing of FOF with MEM and COL was performed using the checkerboard (CB) method. Data were analyzed using logistic regression.</p><p><strong>Results: </strong>Among 50 isolates, the FOF AD MIC<sub>50/90</sub> were 64/>128 μg/mL. Positive in vitro interactions (synergistic or additive) were observed in 43.5% of isolates for both FOF+MEM and FOF+COL, with no antagonism. The clinical and microbiological response rates were 61.5% and 80.5%, 14- and 30-day mortality rates were 21.2% and 40.4%, respectively. Patients receiving at least one agent with in vitro positive interaction had higher clinical response, but the difference wasn't significant (73.7% vs. 52.2% p=0.153). In multivariable analysis, increased FOF AD MIC and Pitt bacteremia score (PBS) were associated with clinical nonresponse, while higher COL MIC and PBS predicted 30-day mortality.</p><p><strong>Conclusion: </strong>FOF AD MIC value may have potential as a predictive marker for FOF-C treatment response in CP-Kp BSI and could aid in guiding combination therapy decisions. Larger, prospective studies using standardized synergy testing methods are needed.</p>","PeriodicalId":13818,"journal":{"name":"International Journal of Antimicrobial Agents","volume":" ","pages":"107687"},"PeriodicalIF":4.6,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145696113","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}
Pub Date : 2025-12-01DOI: 10.1016/j.ijantimicag.2025.107684
Boast A, Legg A, McCarthy J, Roberts Ja, Tong Syc, Duffull S, Gwee A
Background: Ibuprofen is frequently co-administered with antibiotics for its analgesic effect when treating infections. In vitro studies have shown ibuprofen exhibits concentration-dependent inhibition of the organic anion transporters (OAT) 1 and OAT3 and therefore may reduce the clearance of β-lactams including flucloxacillin, in a similar manner to probenecid. We therefore aimed to evaluate the effect of ibuprofen on the pharmacokinetics (PK) of flucloxacillin in healthy volunteers.
Methods: Single site, PK study of 10 healthy adult volunteers. Over a three-day intervention period, participants received intravenous (IV) flucloxacillin 2000 mg twice daily with oral ibuprofen 400 mg twice daily on days 2 and 3. Total and unbound flucloxacillin plasma concentrations were collected at predefined time points. A population PK model was developed using a non-linear mixed effects modelling approach and ibuprofen assessed as a covariate in the final model.
Results: Ten participants with a median age of 24.4 years (range 18.8-54.5) and weight of 68.3 kilograms (range 50.5-85.7) were included. A median of 51.5 (range 34-52) samples were collected per participant. The final two-compartment model included saturable protein binding and allometric scaling of weight on clearance. Ibuprofen was not found to be a significant covariate on Kd (binding dissociation constant), Bmax (maximum binding capacity) or clearance.
Conclusion: Orally administered ibuprofen had no measurable effect on plasma flucloxacillin PK in healthy adults. Given the lack of drug-drug interaction, there is no benefit to prescribe ibuprofen alongside flucloxacillin to increase flucloxacillin exposure.
{"title":"Effect of ibuprofen on the pharmacokinetics of intravenous flucloxacillin in healthy adults.","authors":"Boast A, Legg A, McCarthy J, Roberts Ja, Tong Syc, Duffull S, Gwee A","doi":"10.1016/j.ijantimicag.2025.107684","DOIUrl":"https://doi.org/10.1016/j.ijantimicag.2025.107684","url":null,"abstract":"<p><strong>Background: </strong>Ibuprofen is frequently co-administered with antibiotics for its analgesic effect when treating infections. In vitro studies have shown ibuprofen exhibits concentration-dependent inhibition of the organic anion transporters (OAT) 1 and OAT3 and therefore may reduce the clearance of β-lactams including flucloxacillin, in a similar manner to probenecid. We therefore aimed to evaluate the effect of ibuprofen on the pharmacokinetics (PK) of flucloxacillin in healthy volunteers.</p><p><strong>Methods: </strong>Single site, PK study of 10 healthy adult volunteers. Over a three-day intervention period, participants received intravenous (IV) flucloxacillin 2000 mg twice daily with oral ibuprofen 400 mg twice daily on days 2 and 3. Total and unbound flucloxacillin plasma concentrations were collected at predefined time points. A population PK model was developed using a non-linear mixed effects modelling approach and ibuprofen assessed as a covariate in the final model.</p><p><strong>Results: </strong>Ten participants with a median age of 24.4 years (range 18.8-54.5) and weight of 68.3 kilograms (range 50.5-85.7) were included. A median of 51.5 (range 34-52) samples were collected per participant. The final two-compartment model included saturable protein binding and allometric scaling of weight on clearance. Ibuprofen was not found to be a significant covariate on K<sub>d</sub> (binding dissociation constant), B<sub>max</sub> (maximum binding capacity) or clearance.</p><p><strong>Conclusion: </strong>Orally administered ibuprofen had no measurable effect on plasma flucloxacillin PK in healthy adults. Given the lack of drug-drug interaction, there is no benefit to prescribe ibuprofen alongside flucloxacillin to increase flucloxacillin exposure.</p>","PeriodicalId":13818,"journal":{"name":"International Journal of Antimicrobial Agents","volume":" ","pages":"107684"},"PeriodicalIF":4.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145668231","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}
Pub Date : 2025-12-01DOI: 10.1016/j.ijantimicag.2025.107631
Shuji Gao , Yingying Quan , Shenao Song , Wenjie Jin , Zhiheng Chang , Baobao Liu , Yuxin Wang , Li Yi , Yang Wang
{"title":"Corrigendum to “Disrupting bacterial metabolism by targeting LDH reverses Streptococcus suis aminoglycoside resistance” [International Journal of Antimicrobial Agents 66/6 (2025) 1-16/107599]","authors":"Shuji Gao , Yingying Quan , Shenao Song , Wenjie Jin , Zhiheng Chang , Baobao Liu , Yuxin Wang , Li Yi , Yang Wang","doi":"10.1016/j.ijantimicag.2025.107631","DOIUrl":"10.1016/j.ijantimicag.2025.107631","url":null,"abstract":"","PeriodicalId":13818,"journal":{"name":"International Journal of Antimicrobial Agents","volume":"66 6","pages":"Article 107631"},"PeriodicalIF":4.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145274431","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}
Pub Date : 2025-12-01DOI: 10.1016/j.ijantimicag.2025.107682
Yuxuan Liu, Hanxu Hong, QianBin Dai, Linping Fan, Peng Liu, DanDan Wei, Yang Liu
Objectives: Cefiderocol (FDC) is a promising treatment for infections caused by carbapenem-resistant Klebsiella pneumoniae (CRKP), but emerging heteroresistance (HR) may compromise its clinical efficacy. This study aimed to elucidate the molecular mechanisms underlying FDC heteroresistance arising during in vivo evolution of CRKP. in vivo-evolved.
Methods: A total of 477 clinical CRKP isolates were analyzed. FDC-heteroresistant (FDC-HR) subpopulations emerging during in vivo evolution were characterized using population analysis profiling, antimicrobial susceptibility testing, quantitative PCR, plasmid copy number analysis, transcriptomics, whole-genome sequencing, molecular modeling, and CRISPR-Cas9-based functional validation.
Results: Among the 477 CRKP isolates, 376 (78.8%) belonged to ST11-KL64, 4 (0.84%) were non-susceptible to FDC, and 12 (2.52%) exhibited an FDC-HR phenotype. Resistant subpopulations showed approximately a twofold increase in blaKPC-2 plasmid copy number with upregulated expression compared with susceptible counterparts. A G652A point mutation in mrcA was identified and predicted to impair FDC binding. Functional validation demonstrated that the mrcA mutation alone increased the FDC minimum inhibitory concentration (MIC), while its combination with blaKPC-2 amplification elevated the MIC from 2 to 32 mg/L, fully recapitulating the clinical heteroresistance phenotype. In vivo evolution reduced virulence without affecting growth.
Conclusions: Amplification of blaKPC-2 plasmid copy number together with the mrcA G652A mutation drives FDC-HR in CRKP during in vivo evolution. This mechanism may lead to underestimation of resistance by standard susceptibility testing and increase the risk of treatment failure.
{"title":"bla<sub>KPC-2</sub> amplification and a novel mrcA mutation drive Cefiderocol heteroresistance in ST11 CRKP.","authors":"Yuxuan Liu, Hanxu Hong, QianBin Dai, Linping Fan, Peng Liu, DanDan Wei, Yang Liu","doi":"10.1016/j.ijantimicag.2025.107682","DOIUrl":"10.1016/j.ijantimicag.2025.107682","url":null,"abstract":"<p><strong>Objectives: </strong>Cefiderocol (FDC) is a promising treatment for infections caused by carbapenem-resistant Klebsiella pneumoniae (CRKP), but emerging heteroresistance (HR) may compromise its clinical efficacy. This study aimed to elucidate the molecular mechanisms underlying FDC heteroresistance arising during in vivo evolution of CRKP. in vivo-evolved.</p><p><strong>Methods: </strong>A total of 477 clinical CRKP isolates were analyzed. FDC-heteroresistant (FDC-HR) subpopulations emerging during in vivo evolution were characterized using population analysis profiling, antimicrobial susceptibility testing, quantitative PCR, plasmid copy number analysis, transcriptomics, whole-genome sequencing, molecular modeling, and CRISPR-Cas9-based functional validation.</p><p><strong>Results: </strong>Among the 477 CRKP isolates, 376 (78.8%) belonged to ST11-KL64, 4 (0.84%) were non-susceptible to FDC, and 12 (2.52%) exhibited an FDC-HR phenotype. Resistant subpopulations showed approximately a twofold increase in bla<sub>KPC-2</sub> plasmid copy number with upregulated expression compared with susceptible counterparts. A G652A point mutation in mrcA was identified and predicted to impair FDC binding. Functional validation demonstrated that the mrcA mutation alone increased the FDC minimum inhibitory concentration (MIC), while its combination with bla<sub>KPC-2</sub> amplification elevated the MIC from 2 to 32 mg/L, fully recapitulating the clinical heteroresistance phenotype. In vivo evolution reduced virulence without affecting growth.</p><p><strong>Conclusions: </strong>Amplification of bla<sub>KPC-2</sub> plasmid copy number together with the mrcA G652A mutation drives FDC-HR in CRKP during in vivo evolution. This mechanism may lead to underestimation of resistance by standard susceptibility testing and increase the risk of treatment failure.</p>","PeriodicalId":13818,"journal":{"name":"International Journal of Antimicrobial Agents","volume":" ","pages":"107682"},"PeriodicalIF":4.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145668191","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}
Pub Date : 2025-12-01DOI: 10.1016/j.ijantimicag.2025.107685
Yanchun Qin, Wen Ting Zhou, Ling Wang, Shengqiang Yang, Tao Zhu, Gang Lu, Kwanjit Duangsonk, Hong Zeng
Objective: We aimed to identify novel inhibitors against carbapenem-resistant Acinetobacter baumannii (CR-AB) derived from Streptomyces taklimakanensis sp. nov. TRM43335 and to evaluate their dual antibiofilm and anti-inflammatory mechanisms.
Methods: The TRM43335 extract yielded 3-indole carbaldehyde through bioactivity-guided isolation. Its antibiofilm activity was assessed using crystal violet, extracellular polymeric substances (EPS) quantification, motility, confocal laser scanning microscopy (CLSM), reverse transcription quantitative polymerase chain reaction (RT-qPCR) targeting ompA (outer-membrane protein A), bfmR (biofilm-associated regulator), and bap (biofilm-associated protein), and molecular docking. In vivo efficacy was evaluated in a murine pneumonia model by measuring bacterial burden, histology, and expression of inflammation-related proteins (Toll-like receptor 4 [TLR4], nuclear factor-κB [NF-κB], NOD-like receptor family pyrin domain-containing 3 [NLRP3], and mitogen-activated protein kinase [MAPK] pathway components) via western blotting.
Results: 3-Indole carbaldehyde significantly reduced CR-AB biofilm, suppressed EPS/extracellular proteins, and impaired swarming motility, with marked downregulation of biofilm genes. In mice, 3-indole carbaldehyde treatment decreased lung bacterial load, improved histopathology and inhibited expression of TLR4, NF-κB, NLRP3, and MAPK pathway components (including p38, extracellular signal-regulated kinase 2 [ERK2], c-Jun N-terminal kinase [JNK], tumor necrosis factor receptor-associated factor 6 [TRAF6], and tumor necrosis factor alpha [TNF-α]), indicating suppression of inflammasome and MAPK signaling.
Conclusion: 3-indole carbaldehyde simultaneously inhibits CR-AB biofilm formation and attenuates pulmonary inflammation. Thus, it represents a novel dual-action therapeutic strategy that addresses the key limitations of single-target antibiotics such as meropenem. Coupled with a favorable safety profile, it is a highly promising candidate for treating drug-resistant CR-AB infections.
目的:鉴定塔克拉玛卡纳链霉菌TRM43335中抗碳青霉烯耐药鲍曼不动杆菌(CR-AB)的新型抑制剂,并评价其双抗膜和抗炎机制。方法:TRM43335提取物通过生物活性引导分离得到3-吲哚醛。采用结晶紫、细胞外聚合物质(EPS)定量、运动、共聚焦激光扫描显微镜(CLSM)、针对膜外蛋白A (ompA)、生物膜相关调节因子bfmR和生物膜相关蛋白bap的逆转录定量聚合酶链反应(RT-qPCR)和分子对接等方法评估其抗生物膜活性。在小鼠肺炎模型中,通过免疫印迹法测定细菌负荷、组织学和炎症相关蛋白(toll样受体4 [TLR4]、核因子-κB [NF-κB]、nod样受体家族pyrin结构域3 [NLRP3]和丝裂原活化蛋白激酶[MAPK]途径组分)的表达来评估体内疗效。结果:3-吲哚醛显著降低CR-AB生物膜,抑制EPS/细胞外蛋白,损害蜂群运动,生物膜基因明显下调。在小鼠中,3-吲哚醛处理降低了肺部细菌负荷,改善了组织病理学,抑制了TLR4、NF-κB、NLRP3和MAPK通路组分(包括p38、细胞外信号调节激酶2 [ERK2]、c-Jun n -末端激酶[JNK]、肿瘤坏死因子受体相关因子6 [TRAF6]和肿瘤坏死因子α [TNF-α])的表达,表明炎症小体和MAPK信号传导受到抑制。结论:3-吲哚乙醛同时抑制CR-AB生物膜的形成,减轻肺部炎症。因此,它代表了一种新的双作用治疗策略,解决了单靶点抗生素(如美罗培南)的主要局限性。再加上良好的安全性,它是治疗耐药CR-AB感染的极有希望的候选药物。
{"title":"Streptomyces taklimakanensis sp. nov. TRM43335 produces 3-indole carbaldehyde a promising inhibitor of carbapenem-resistant Acinetobacter baumannii: in vitro and in vivo evaluation.","authors":"Yanchun Qin, Wen Ting Zhou, Ling Wang, Shengqiang Yang, Tao Zhu, Gang Lu, Kwanjit Duangsonk, Hong Zeng","doi":"10.1016/j.ijantimicag.2025.107685","DOIUrl":"https://doi.org/10.1016/j.ijantimicag.2025.107685","url":null,"abstract":"<p><strong>Objective: </strong>We aimed to identify novel inhibitors against carbapenem-resistant Acinetobacter baumannii (CR-AB) derived from Streptomyces taklimakanensis sp. nov. TRM43335 and to evaluate their dual antibiofilm and anti-inflammatory mechanisms.</p><p><strong>Methods: </strong>The TRM43335 extract yielded 3-indole carbaldehyde through bioactivity-guided isolation. Its antibiofilm activity was assessed using crystal violet, extracellular polymeric substances (EPS) quantification, motility, confocal laser scanning microscopy (CLSM), reverse transcription quantitative polymerase chain reaction (RT-qPCR) targeting ompA (outer-membrane protein A), bfmR (biofilm-associated regulator), and bap (biofilm-associated protein), and molecular docking. In vivo efficacy was evaluated in a murine pneumonia model by measuring bacterial burden, histology, and expression of inflammation-related proteins (Toll-like receptor 4 [TLR4], nuclear factor-κB [NF-κB], NOD-like receptor family pyrin domain-containing 3 [NLRP3], and mitogen-activated protein kinase [MAPK] pathway components) via western blotting.</p><p><strong>Results: </strong>3-Indole carbaldehyde significantly reduced CR-AB biofilm, suppressed EPS/extracellular proteins, and impaired swarming motility, with marked downregulation of biofilm genes. In mice, 3-indole carbaldehyde treatment decreased lung bacterial load, improved histopathology and inhibited expression of TLR4, NF-κB, NLRP3, and MAPK pathway components (including p38, extracellular signal-regulated kinase 2 [ERK2], c-Jun N-terminal kinase [JNK], tumor necrosis factor receptor-associated factor 6 [TRAF6], and tumor necrosis factor alpha [TNF-α]), indicating suppression of inflammasome and MAPK signaling.</p><p><strong>Conclusion: </strong>3-indole carbaldehyde simultaneously inhibits CR-AB biofilm formation and attenuates pulmonary inflammation. Thus, it represents a novel dual-action therapeutic strategy that addresses the key limitations of single-target antibiotics such as meropenem. Coupled with a favorable safety profile, it is a highly promising candidate for treating drug-resistant CR-AB infections.</p>","PeriodicalId":13818,"journal":{"name":"International Journal of Antimicrobial Agents","volume":" ","pages":"107685"},"PeriodicalIF":4.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145668246","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}