Pub Date : 2025-02-13Epub Date: 2024-12-17DOI: 10.1128/aac.01377-24
Cuiyun Li, Jiajia Mai, Min Wu, Hong Zhang, Xiaojiao Li, Haijun Li, Youyun Li, Yanhua Ding
This study evaluated the safety and pharmacokinetics (PK) of a single dose of leritrelvir, a novel inhibitor of 3-chymotrypsin-like cysteine protease (3CLpro), in patients with hepatic impairment versus healthy participants with normal hepatic function. Eight participants with mild (Child-Pugh A) hepatic impairment, eight with moderate (Child-Pugh B) hepatic impairment, and eight healthy matched control participants were enrolled in this open-label, parallel clinical trial. After administration of leritrelvir of 400 mg, PK parameters were calculated and compared across groups. In total, 24 participants were enrolled and completed the study. Leritrelvir was generally well tolerated, with no serious adverse events or deaths reported during the study. Compared to the group with normal hepatic function, the geometric least-squares mean ratios (90% confidence intervals) for Cmax, AUC0-t, and AUC0-∞ of leritrelvir in participants with mild hepatic impairment were 96.9% (69.3%, 135%), 92.2% (69.6%, 122%), and 92.1% (69.7%, 122%), respectively. For moderate hepatic impairment, the corresponding ratios were 91.6% (61.7%, 136%), 113% (80.0%, 160%), and 113% (80.0%, 159%). Leritrelvir exposures were comparable among the three groups. Overall, there was no clinically relevant difference in leritrelvir exposure in participants with hepatic impairment compared to normal controls. No dose adjustment is required for leritrelvir in patients with mild or moderate hepatic impairment.CLINICAL TRIALSThis study is registered with ClinicalTrials.gov as NCT06161259.
{"title":"Single-dose tolerability and pharmacokinetics of leritrelvir in Chinese patients with hepatic impairment and healthy matched controls.","authors":"Cuiyun Li, Jiajia Mai, Min Wu, Hong Zhang, Xiaojiao Li, Haijun Li, Youyun Li, Yanhua Ding","doi":"10.1128/aac.01377-24","DOIUrl":"10.1128/aac.01377-24","url":null,"abstract":"<p><p>This study evaluated the safety and pharmacokinetics (PK) of a single dose of leritrelvir, a novel inhibitor of 3-chymotrypsin-like cysteine protease (3CLpro), in patients with hepatic impairment versus healthy participants with normal hepatic function. Eight participants with mild (Child-Pugh A) hepatic impairment, eight with moderate (Child-Pugh B) hepatic impairment, and eight healthy matched control participants were enrolled in this open-label, parallel clinical trial. After administration of leritrelvir of 400 mg, PK parameters were calculated and compared across groups. In total, 24 participants were enrolled and completed the study. Leritrelvir was generally well tolerated, with no serious adverse events or deaths reported during the study. Compared to the group with normal hepatic function, the geometric least-squares mean ratios (90% confidence intervals) for <i>C</i><sub>max</sub>, AUC<sub>0-<i>t</i></sub>, and AUC<sub>0-∞</sub> of leritrelvir in participants with mild hepatic impairment were 96.9% (69.3%, 135%), 92.2% (69.6%, 122%), and 92.1% (69.7%, 122%), respectively. For moderate hepatic impairment, the corresponding ratios were 91.6% (61.7%, 136%), 113% (80.0%, 160%), and 113% (80.0%, 159%). Leritrelvir exposures were comparable among the three groups. Overall, there was no clinically relevant difference in leritrelvir exposure in participants with hepatic impairment compared to normal controls. No dose adjustment is required for leritrelvir in patients with mild or moderate hepatic impairment.CLINICAL TRIALSThis study is registered with ClinicalTrials.gov as NCT06161259.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0137724"},"PeriodicalIF":4.1,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11823647/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142833524","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-13Epub Date: 2025-01-10DOI: 10.1128/aac.01040-24
Dominic M H Tong, Maria-Stephanie A Hughes, Jasmine Hu, Jeffrey C Pearson, David W Kubiak, Brandon W Dionne, Jasmine H Hughes
Tobramycin dosing in patients with cystic fibrosis (CF) is challenged by its high pharmacokinetic (PK) variability and narrow therapeutic window. Doses are typically individualized using two-sample log-linear regression (LLR) to quantify the area under the concentration-time curve (AUC). Bayesian model-informed precision dosing (MIPD) may allow dose individualization with fewer samples; however, the relative performance of these methods is unknown. This single-center retrospective analysis included adult patients with CF receiving tobramycin from 2015 to 2022. Tobramycin concentrations were predicted using LLR or Bayesian estimation with two population PK models (Hennig and Alghanem). Then, both methods were used to estimate the AUC for simulated patients. For Bayesian estimation, AUC estimation with flattened priors and limited sampling strategies were also assessed. Predictions were evaluated using normalized root mean square error (nRMSE), mean percent error (MPE), and accuracy. The data set included 70 treatment courses, with 32 not evaluable by LLR due to detection limits or timing issues. Bayesian estimation demonstrated worse accuracy (47.1%-50.7% vs 75.7%), higher MPE (24.2%-32.4% vs -2.4%), and higher nRMSE (35.0%-39.4% vs 24.8%) than LLR for peak concentrations but performed better on troughs (accuracy: 92.0%-92.9% vs 84.6%). Bayesian estimation with flattened priors and a single sample at 4 h was comparable to LLR performance, with better accuracy (42.9%-68.0% vs 41.1% LLR), comparable MPE (-2.3% to -3.7% vs -0.5%) and nRMSE (11.3%-21.6% vs 17.3%). Bayesian estimation with one concentration and flattened priors can match LLR prediction accuracy. However, popPK models must be improved to better estimate peak samples.
囊性纤维化(CF)患者的妥布霉素剂量受到其高药代动力学(PK)变异性和狭窄治疗窗口的挑战。剂量通常使用双样本对数线性回归(LLR)来量化浓度-时间曲线(AUC)下的面积。基于贝叶斯模型的精确剂量(MIPD)可以在较少的样本下实现剂量个性化;然而,这些方法的相对性能是未知的。这项单中心回顾性分析纳入了2015年至2022年接受妥布霉素治疗的成年CF患者。妥布霉素浓度预测使用LLR或贝叶斯估计与两个种群PK模型(Hennig和Alghanem)。然后,使用这两种方法估计模拟患者的AUC。对于贝叶斯估计,还评估了平坦先验和有限采样策略下的AUC估计。使用标准化均方根误差(nRMSE)、平均百分比误差(MPE)和准确性评估预测。数据集包括70个疗程,由于检测限制或时间问题,32个疗程无法用LLR评估。与LLR相比,贝叶斯估计对峰值浓度的准确率较低(47.1%-50.7%对75.7%),MPE较高(24.2%-32.4%对-2.4%),nRMSE较高(35.0%-39.4%对24.8%),但对波谷的准确率较好(准确率:92.0%-92.9%对84.6%)。具有平坦先验和4小时单个样本的贝叶斯估计与LLR性能相当,具有更好的准确性(42.9%-68.0% vs 41.1% LLR),可比较的MPE(-2.3%至-3.7% vs -0.5%)和nRMSE (11.3%-21.6% vs 17.3%)。单浓度、平坦先验的贝叶斯估计可以达到LLR的预测精度。然而,popPK模型必须改进才能更好地估计峰值样本。
{"title":"Comparing two-sample log-linear exposure estimation with Bayesian model-informed precision dosing of tobramycin in adult patients with cystic fibrosis.","authors":"Dominic M H Tong, Maria-Stephanie A Hughes, Jasmine Hu, Jeffrey C Pearson, David W Kubiak, Brandon W Dionne, Jasmine H Hughes","doi":"10.1128/aac.01040-24","DOIUrl":"10.1128/aac.01040-24","url":null,"abstract":"<p><p>Tobramycin dosing in patients with cystic fibrosis (CF) is challenged by its high pharmacokinetic (PK) variability and narrow therapeutic window. Doses are typically individualized using two-sample log-linear regression (LLR) to quantify the area under the concentration-time curve (AUC). Bayesian model-informed precision dosing (MIPD) may allow dose individualization with fewer samples; however, the relative performance of these methods is unknown. This single-center retrospective analysis included adult patients with CF receiving tobramycin from 2015 to 2022. Tobramycin concentrations were predicted using LLR or Bayesian estimation with two population PK models (Hennig and Alghanem). Then, both methods were used to estimate the AUC for simulated patients. For Bayesian estimation, AUC estimation with flattened priors and limited sampling strategies were also assessed. Predictions were evaluated using normalized root mean square error (nRMSE), mean percent error (MPE), and accuracy. The data set included 70 treatment courses, with 32 not evaluable by LLR due to detection limits or timing issues. Bayesian estimation demonstrated worse accuracy (47.1%-50.7% vs 75.7%), higher MPE (24.2%-32.4% vs -2.4%), and higher nRMSE (35.0%-39.4% vs 24.8%) than LLR for peak concentrations but performed better on troughs (accuracy: 92.0%-92.9% vs 84.6%). Bayesian estimation with flattened priors and a single sample at 4 h was comparable to LLR performance, with better accuracy (42.9%-68.0% vs 41.1% LLR), comparable MPE (-2.3% to -3.7% vs -0.5%) and nRMSE (11.3%-21.6% vs 17.3%). Bayesian estimation with one concentration and flattened priors can match LLR prediction accuracy. However, popPK models must be improved to better estimate peak samples.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0104024"},"PeriodicalIF":4.1,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11823644/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142943340","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-13Epub Date: 2024-12-19DOI: 10.1128/aac.00739-24
Charles O Obonyo, Vincent O Were, Peter Wamae, Erick M O Muok
Praziquantel alone is insufficient for the control of schistosomiasis due to poor efficacy against juvenile worms and increasing concerns about the risk of drug resistance. We compared the efficacy and safety of praziquantel combined with four different artemisinin-based combinations to praziquantel alone in treating Schistosoma mansoni infection in Kenyan children. In this randomized, open-label, five-arm, head-to-head, non-inferiority trial, children (aged 9-15 years) with S. mansoni infection according to duplicate Kato Katz thick smears from a stool sample in the Mwea irrigation scheme of central Kenya, were enrolled. Participants were randomly assigned (1:1:1:1:1) via a computer-generated block randomization procedure to receive a single oral dose of praziquantel (PZQ) (40 mg/kg/day) alone or in combination with a 3-day course (4 mg/kg of artesunate) of artesunate plus sulfalene-pyrimethamine (As + SP), artesunate plus amodiaquine (As + AQ), artesunate plus mefloquine (As + MQ) or dihydroartemisinin-piperaquine (DHAP). Laboratory technicians were masked to treatment allocation, but participants, clinicians, and study nurses were not. The primary outcomes were the cure rate and frequency of adverse events, which were assessed 6 weeks after treatment in the available case population using a per-protocol analysis. The non-inferiority margin was set at -10% for the risk difference in cure rates between combination therapy and PZQ alone. Between 12 September 2018 and 11 January 2019, 540 participants were assigned to receive PZQ alone (n = 108), PZQ plus As + SP (n = 108), PZQ plus As + AQ (n = 108), PZQ plus As + MQ (n = 108), or PZQ plus DHAP (n = 108). Primary outcome data were available for 523 (96.9%) participants. The cure rate was 82.5% (85/103) in PZQ alone, 81.7% (85/104) in PZQ plus As + SP, 76.2% (80/105) in PZQ plus As + AQ, 88.7% (94/106) in PZQ plus As + MQ, and 85.7% (90/105) in PZQ plus DHAP arm. Non-inferiority was declared for PZQ plus As + MQ (difference 6.2 [95% confidence interval: -3.3 to 15.6]) and PZQ plus DHAP (3.2 [-6.7 to 13.1]) but not for PZQ plus As + SP (-0.8 [-11.2 to 9.6]) or PZQ plus As + AQ (-6.3 [-17.3 to 4.6]). Adverse events were reported by 26% (138/540) of participants, including abdominal pain, headache, and vomiting. There were no serious adverse events. Alternatives to praziquantel should include praziquantel plus artesunate-mefloquine or praziquantel plus dihydroartemisinin-piperaquine. However, further multicentre trials are needed in different epidemiological settings and population groups to confirm these findings.CLINICAL TRIALSThis study is registered with the Pan-African Clinical Trials Registry under PACTR202001919442161.
单独使用吡喹酮不足以控制血吸虫病,因为它对血吸虫幼虫的疗效较差,而且对耐药风险的担忧日益增加。我们比较了吡喹酮联合四种不同的以青蒿素为基础的组合与吡喹酮单独治疗肯尼亚儿童曼氏血吸虫感染的疗效和安全性。在这项随机、开放标签、五组、头对头、非劣效性试验中,根据肯尼亚中部Mwea灌溉计划中粪便样本的重复Kato Katz厚涂片,纳入了9-15岁的曼氏链球菌感染儿童。参与者通过计算机生成的块随机程序随机分配(1:1:1:1:1),接受单次口服吡喹酮(PZQ) (40 mg/kg/天)或与3天疗程(4 mg/kg青蒿琥酯)的青蒿琥酯加磺胺-乙胺嘧啶(As + SP),青蒿琥酯加阿莫地喹(As + AQ),青蒿琥酯加甲氟喹(As + MQ)或双氢青蒿素-哌喹(DHAP)联合使用。实验室技术人员不知道治疗分配情况,但参与者、临床医生和研究护士不知道。主要结果是治愈率和不良事件发生的频率,在治疗后6周对现有病例群体进行评估,使用每个方案分析。联合治疗和单独使用PZQ的治愈率风险差异的非劣效性边际设定为-10%。在2018年9月12日至2019年1月11日期间,540名参与者被分配接受单独PZQ (n = 108), PZQ加As + SP (n = 108), PZQ加As + AQ (n = 108), PZQ加As + MQ (n = 108)或PZQ加DHAP (n = 108)。523名(96.9%)参与者可获得主要结局数据。单纯PZQ组治愈率为82.5% (85/103),PZQ + As + SP组治愈率为81.7% (85/104),PZQ + As + AQ组治愈率为76.2% (80/105),PZQ + As + MQ组治愈率为88.7% (94/106),PZQ + DHAP组治愈率为85.7%(90/105)。PZQ + As + MQ(差异为6.2[95%置信区间:-3.3至15.6])和PZQ + DHAP(差异为3.2[-6.7至13.1])非劣效性,但PZQ + As + SP(-0.8[-11.2至9.6])或PZQ + As + AQ(-6.3[-17.3至4.6])不具有劣效性。26%(138/540)的参与者报告了不良事件,包括腹痛、头痛和呕吐。无严重不良事件发生。吡喹酮的替代品应包括吡喹酮加青蒿琥酯-甲氟喹或吡喹酮加双氢青蒿素-哌喹。然而,需要在不同的流行病学环境和人群中进行进一步的多中心试验来证实这些发现。临床试验本研究已在泛非临床试验注册中心注册,注册号为PACTR202001919442161。
{"title":"Efficacy and safety of praziquantel plus artemisinin-based combinations versus praziquantel in the treatment of Kenyan children with <i>Schistosoma mansoni</i> infection: open-label, randomized, head-to-head, non-inferiority trial.","authors":"Charles O Obonyo, Vincent O Were, Peter Wamae, Erick M O Muok","doi":"10.1128/aac.00739-24","DOIUrl":"10.1128/aac.00739-24","url":null,"abstract":"<p><p>Praziquantel alone is insufficient for the control of schistosomiasis due to poor efficacy against juvenile worms and increasing concerns about the risk of drug resistance. We compared the efficacy and safety of praziquantel combined with four different artemisinin-based combinations to praziquantel alone in treating <i>Schistosoma mansoni</i> infection in Kenyan children. In this randomized, open-label, five-arm, head-to-head, non-inferiority trial, children (aged 9-15 years) with <i>S. mansoni</i> infection according to duplicate Kato Katz thick smears from a stool sample in the Mwea irrigation scheme of central Kenya, were enrolled. Participants were randomly assigned (1:1:1:1:1) via a computer-generated block randomization procedure to receive a single oral dose of praziquantel (PZQ) (40 mg/kg/day) alone or in combination with a 3-day course (4 mg/kg of artesunate) of artesunate plus sulfalene-pyrimethamine (As + SP), artesunate plus amodiaquine (As + AQ), artesunate plus mefloquine (As + MQ) or dihydroartemisinin-piperaquine (DHAP). Laboratory technicians were masked to treatment allocation, but participants, clinicians, and study nurses were not. The primary outcomes were the cure rate and frequency of adverse events, which were assessed 6 weeks after treatment in the available case population using a per-protocol analysis. The non-inferiority margin was set at -10% for the risk difference in cure rates between combination therapy and PZQ alone. Between 12 September 2018 and 11 January 2019, 540 participants were assigned to receive PZQ alone (<i>n</i> = 108), PZQ plus As + SP (<i>n</i> = 108), PZQ plus As + AQ (<i>n</i> = 108), PZQ plus As + MQ (<i>n</i> = 108), or PZQ plus DHAP (<i>n</i> = 108). Primary outcome data were available for 523 (96.9%) participants. The cure rate was 82.5% (85/103) in PZQ alone, 81.7% (85/104) in PZQ plus As + SP, 76.2% (80/105) in PZQ plus As + AQ, 88.7% (94/106) in PZQ plus As + MQ, and 85.7% (90/105) in PZQ plus DHAP arm. Non-inferiority was declared for PZQ plus As + MQ (difference 6.2 [95% confidence interval: -3.3 to 15.6]) and PZQ plus DHAP (3.2 [-6.7 to 13.1]) but not for PZQ plus As + SP (-0.8 [-11.2 to 9.6]) or PZQ plus As + AQ (-6.3 [-17.3 to 4.6]). Adverse events were reported by 26% (138/540) of participants, including abdominal pain, headache, and vomiting. There were no serious adverse events. Alternatives to praziquantel should include praziquantel plus artesunate-mefloquine or praziquantel plus dihydroartemisinin-piperaquine. However, further multicentre trials are needed in different epidemiological settings and population groups to confirm these findings.CLINICAL TRIALSThis study is registered with the Pan-African Clinical Trials Registry under PACTR202001919442161.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0073924"},"PeriodicalIF":4.1,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11823657/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142852236","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-13Epub Date: 2025-01-17DOI: 10.1128/aac.01251-24
Jinhee Jo, Chenlin Hu, Thomas D Horvath, Sigmund J Haidacher, Khurshida Begum, M Jahangir Alam, Kevin W Garey
Omadacycline, an aminomethylcycline tetracycline, has a low propensity to cause Clostridioides difficile infection (CDI) in clinical trials. Omadacycline exhibited a reduced bactericidal effect compared with vancomycin on key microorganisms implicated in bile acid homeostasis and short-chain fatty acids (SCFAs), key components of CDI pathogenesis. The purpose of this study was to assess bile acid and SCFA changes in stool samples from healthy volunteers given omadacycline or vancomycin. Stool samples were collected daily from 16 healthy volunteers, who were given oral omadacycline or vancomycin for 10 days. Daily stool samples were assessed for bile acids and SCFA concentrations using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Bile acids changed significantly over time for all subjects (P < 0.01 for each bile acid), with vancomycin causing a larger change in the primary bile acids, cholic acid (P < 0.001) and chenodeoxycholic acid (P < 0.001), and a reduced change in the secondary bile acid, lithocholic acid (P < 0.001). The secondary bile acid ursodeoxycholic acid was reduced less by vancomycin than by omadacycline (P < 0.001). All SCFA concentrations were reduced from baseline with a larger effect observed with vancomycin for isobutyric acid (P = 0.0034), propionic acid (P = 0.0012), and acetic acid (P = 0.047). Microbial changes associated with the use of vancomycin versus omadacycline were also associated with changes in bile acid homeostasis and SCFA concentrations. Oral omadacycline produced a distinctive metabolomic profile compared with vancomycin when administered to healthy subjects. The metabolic findings help further our understanding of the lower CDI risk properties of omadacycline and warrant phase 2 investigations using omadacycline as a CDI antibiotic.
Importance: The purpose of this study was to assess bile acid and SCFA changes in stool samples obtained from healthy volunteers given omadacycline or vancomycin. Stool samples were collected daily from 16 healthy volunteers given a 10-day oral course of omadacycline or vancomycin. Vancomycin caused a larger change in the primary bile acids and SCFA concentrations compared with omadacycline. The metabolic findings help further our understanding of the mechanistic basis for the lower-risk properties of omadacycline causing CDI and warrant phase 2 investigations using omadacycline as a CDI antibiotic.
Clinical trials: This study is registered with ClinicalTrials.gov as NCT06030219.
{"title":"Phase I trial comparing bile acid and short-chain fatty acid alterations in stool collected from human subjects treated with omadacycline or vancomycin.","authors":"Jinhee Jo, Chenlin Hu, Thomas D Horvath, Sigmund J Haidacher, Khurshida Begum, M Jahangir Alam, Kevin W Garey","doi":"10.1128/aac.01251-24","DOIUrl":"10.1128/aac.01251-24","url":null,"abstract":"<p><p>Omadacycline, an aminomethylcycline tetracycline, has a low propensity to cause <i>Clostridioides difficile</i> infection (CDI) in clinical trials. Omadacycline exhibited a reduced bactericidal effect compared with vancomycin on key microorganisms implicated in bile acid homeostasis and short-chain fatty acids (SCFAs), key components of CDI pathogenesis. The purpose of this study was to assess bile acid and SCFA changes in stool samples from healthy volunteers given omadacycline or vancomycin. Stool samples were collected daily from 16 healthy volunteers, who were given oral omadacycline or vancomycin for 10 days. Daily stool samples were assessed for bile acids and SCFA concentrations using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Bile acids changed significantly over time for all subjects (<i>P</i> < 0.01 for each bile acid), with vancomycin causing a larger change in the primary bile acids, cholic acid (<i>P</i> < 0.001) and chenodeoxycholic acid (<i>P</i> < 0.001), and a reduced change in the secondary bile acid, lithocholic acid (<i>P</i> < 0.001). The secondary bile acid ursodeoxycholic acid was reduced less by vancomycin than by omadacycline (<i>P</i> < 0.001). All SCFA concentrations were reduced from baseline with a larger effect observed with vancomycin for isobutyric acid (<i>P</i> = 0.0034), propionic acid (<i>P</i> = 0.0012), and acetic acid (<i>P</i> = 0.047). Microbial changes associated with the use of vancomycin versus omadacycline were also associated with changes in bile acid homeostasis and SCFA concentrations. Oral omadacycline produced a distinctive metabolomic profile compared with vancomycin when administered to healthy subjects. The metabolic findings help further our understanding of the lower CDI risk properties of omadacycline and warrant phase 2 investigations using omadacycline as a CDI antibiotic.</p><p><strong>Importance: </strong>The purpose of this study was to assess bile acid and SCFA changes in stool samples obtained from healthy volunteers given omadacycline or vancomycin. Stool samples were collected daily from 16 healthy volunteers given a 10-day oral course of omadacycline or vancomycin. Vancomycin caused a larger change in the primary bile acids and SCFA concentrations compared with omadacycline. The metabolic findings help further our understanding of the mechanistic basis for the lower-risk properties of omadacycline causing CDI and warrant phase 2 investigations using omadacycline as a CDI antibiotic.</p><p><strong>Clinical trials: </strong>This study is registered with ClinicalTrials.gov as NCT06030219.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0125124"},"PeriodicalIF":4.1,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11823362/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142999072","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-13Epub Date: 2024-12-23DOI: 10.1128/aac.01200-24
Bonginkosi Ndzamba, Paolo Denti, Helen McIlleron, Peter Smith, Thuli Mthiyane, Roxana Rustomjee, Philip Onyebujoh, Juan Eduardo Reséndiz-Galván
Ethambutol is used to treat tuberculosis (TB) in individuals living with HIV. Low concentrations of ethambutol have been reported in patients dosed with the World Health Organization (WHO)-recommended first-line regimen. We analyzed the pharmacokinetics of ethambutol in 61 HIV-positive individuals diagnosed with drug-sensitive TB enrolled in the tuberculosis and highly active antiretroviral therapy (TB-HAART) study. Participants started on TB treatment and were randomized to early or later introduction of efavirenz-based antiretroviral treatment. We explored potential covariate effects and evaluated the current WHO dosing recommendations for ethambutol in drug-susceptible and multidrug-resistant (MDR)-TB. A two-compartment model with first-order elimination allometrically scaled by fat-free mass and transit compartment absorption best described the pharmacokinetics of ethambutol. Clearance was estimated to be 40.3 L/h for a typical individual with a fat-free mass (FFM) of 42 kg. The Antib-4 formulation had 26% higher bioavailability and slower mean transit time by 37% compared with Rifafour. Simulations showed that individuals in the lower weight bands (<55 kg) who were administered ethambutol at WHO-recommended doses had relatively low drug exposures. These individuals would need doses of 825 mg if their body weight is <37.9 kg and 1,100 mg if it is between 38 and 54.9 kg to achieve the reference maximum concentrations of 2-6 mg/L and an area under the concentration-time curve (0-24) of 16-29 mg·h/L. To achieve these targets in MDR-TB treatment, a dose increment of 400 mg (extra tablet) would be required for individuals in the lower weight band (<46 kg). Our dose adjustments are consistent with the literature and can be recommended for consideration by the WHO for first-line drug-susceptible and MDR-TB treatment.
{"title":"Pharmacokinetics of ethambutol and weight banded dosing in South African adults newly diagnosed with tuberculosis and HIV.","authors":"Bonginkosi Ndzamba, Paolo Denti, Helen McIlleron, Peter Smith, Thuli Mthiyane, Roxana Rustomjee, Philip Onyebujoh, Juan Eduardo Reséndiz-Galván","doi":"10.1128/aac.01200-24","DOIUrl":"10.1128/aac.01200-24","url":null,"abstract":"<p><p>Ethambutol is used to treat tuberculosis (TB) in individuals living with HIV. Low concentrations of ethambutol have been reported in patients dosed with the World Health Organization (WHO)-recommended first-line regimen. We analyzed the pharmacokinetics of ethambutol in 61 HIV-positive individuals diagnosed with drug-sensitive TB enrolled in the tuberculosis and highly active antiretroviral therapy (TB-HAART) study. Participants started on TB treatment and were randomized to early or later introduction of efavirenz-based antiretroviral treatment. We explored potential covariate effects and evaluated the current WHO dosing recommendations for ethambutol in drug-susceptible and multidrug-resistant (MDR)-TB. A two-compartment model with first-order elimination allometrically scaled by fat-free mass and transit compartment absorption best described the pharmacokinetics of ethambutol. Clearance was estimated to be 40.3 L/h for a typical individual with a fat-free mass (FFM) of 42 kg. The Antib-4 formulation had 26% higher bioavailability and slower mean transit time by 37% compared with Rifafour. Simulations showed that individuals in the lower weight bands (<55 kg) who were administered ethambutol at WHO-recommended doses had relatively low drug exposures. These individuals would need doses of 825 mg if their body weight is <37.9 kg and 1,100 mg if it is between 38 and 54.9 kg to achieve the reference maximum concentrations of 2-6 mg/L and an area under the concentration-time curve (0-24) of 16-29 mg·h/L. To achieve these targets in MDR-TB treatment, a dose increment of 400 mg (extra tablet) would be required for individuals in the lower weight band (<46 kg). Our dose adjustments are consistent with the literature and can be recommended for consideration by the WHO for first-line drug-susceptible and MDR-TB treatment.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0120024"},"PeriodicalIF":4.1,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11823665/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142876133","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-13Epub Date: 2024-12-31DOI: 10.1128/aac.01448-24
Roberto Sánchez-Sánchez, Ana Huertas-López, Andrea Largo-de la Torre, Ignacio Ferre, Filippo Maria Dini, Michela Re, Javier Moreno-Gonzalo, Ryan Choi, Matthew A Hulverson, Kayode K Ojo, Samuel L M Arnold, Andrew Hemphill, Wesley C Van Voorhis, Luis Miguel Ortega-Mora
Drug development for congenital toxoplasmosis is challenging since first-line therapy has a high rate of adverse effects and exhibits suboptimal efficacy. Bumped kinase inhibitors (BKIs), targeting protein kinases with small gatekeeper residues, have been found to be effective against Toxoplasma gondii. The efficacy of BKI-1748 administered later than 2 days post-infection (p.i.), a scenario that may better reflect its real-world use as a therapeutic candidate, has not been investigated in T. gondii-infected pregnant sheep. For this purpose, 19 pregnant sheep were assigned to three experimental groups. Group 1 (G1, n = 8) and group 2 (G2, n = 8) were dosed orally with 10 TgShSp1 sporulated oocysts at 90 days of gestation (dg). Animals from group 3 (G3, n = 3) were simultaneously mock dosed with phosphate-buffered solution (PBS). In G1, BKI-1748 was administered orally from day 7 p.i. (fever and increased serum IFNγ levels) onward, maintaining drug exposure for 20 days (10 doses at 15 mg/kg every 2 days). Treated animals (G1) exhibited significantly lower rectal temperatures (on days 8 and 9 p.i.), serum IFNγ levels (on day 10 p.i.), and specific IgG levels when compared with non-treated animals (G2). At delivery, significantly higher percentages of healthy lambs were found in infected/treated sheep in G1 (73.3%) and in uninfected sheep in G3 (80%) compared with infected/untreated sheep in G2 (31.3%). Concerning congenital transmission, parasite DNA was neither detected in placenta nor target tissues (brain and lungs) from the fetuses/lambs in G1(infected/treated) and G3 (uninfected). By contrast, parasite DNA was detected in all placentas and lambs from G2 (infected/untreated), except for one sheep that aborted on day 13 p.i.
先天性弓形虫病的药物开发是具有挑战性的,因为一线治疗有很高的不良反应率,并表现出不理想的疗效。靶向具有小门阀残基的蛋白激酶的碰撞激酶抑制剂(BKIs)已被发现对弓形虫有效。BKI-1748在感染后2天(p.i.)后使用的效果尚未在感染弓形虫的怀孕绵羊中进行调查,这一情况可能更好地反映了其作为候选治疗药物的实际用途。为此,将19只怀孕绵羊分为3个实验组。组1 (G1, n = 8)和组2 (G2, n = 8)在妊娠90天(dg)口服10个TgShSp1孢子卵囊。第三组(G3, n = 3)同时模拟给药磷酸盐缓冲溶液(PBS)。在G1期,BKI-1748从第7天(发烧和血清IFNγ水平升高)开始口服,持续20天(每2天10次,剂量为15 mg/kg)。与未治疗动物(G2)相比,治疗动物(G1)表现出显著降低的直肠温度(第8天和第9天)、血清IFNγ水平(第10天)和特异性IgG水平。分娩时,G1期感染/治疗过的羊的健康羔羊比例(73.3%)和G3期未感染的羊的健康羔羊比例(80%)明显高于G2期感染/未治疗的羊(31.3%)。关于先天性传播,在G1组(感染/治疗组)和G3组(未感染组)的胎儿/羔羊的胎盘和靶组织(脑和肺)中均未检测到寄生虫DNA。相比之下,除了一只在第13天流产的羊外,所有G2(感染/未治疗)的胎盘和羔羊中都检测到寄生虫DNA。
{"title":"Treatment with BKI-1748 after <i>Toxoplasma gondii</i> systemic dissemination in experimentally infected pregnant sheep improves fetal and lamb mortality and morbidity and prevents congenital infection.","authors":"Roberto Sánchez-Sánchez, Ana Huertas-López, Andrea Largo-de la Torre, Ignacio Ferre, Filippo Maria Dini, Michela Re, Javier Moreno-Gonzalo, Ryan Choi, Matthew A Hulverson, Kayode K Ojo, Samuel L M Arnold, Andrew Hemphill, Wesley C Van Voorhis, Luis Miguel Ortega-Mora","doi":"10.1128/aac.01448-24","DOIUrl":"10.1128/aac.01448-24","url":null,"abstract":"<p><p>Drug development for congenital toxoplasmosis is challenging since first-line therapy has a high rate of adverse effects and exhibits suboptimal efficacy. Bumped kinase inhibitors (BKIs), targeting protein kinases with small gatekeeper residues, have been found to be effective against <i>Toxoplasma gondii</i>. The efficacy of BKI-1748 administered later than 2 days post-infection (p.i.), a scenario that may better reflect its real-world use as a therapeutic candidate, has not been investigated in <i>T. gondii</i>-infected pregnant sheep. For this purpose, 19 pregnant sheep were assigned to three experimental groups. Group 1 (G1, <i>n</i> = 8) and group 2 (G2, <i>n</i> = 8) were dosed orally with 10 TgShSp1 sporulated oocysts at 90 days of gestation (dg). Animals from group 3 (G3, <i>n</i> = 3) were simultaneously mock dosed with phosphate-buffered solution (PBS). In G1, BKI-1748 was administered orally from day 7 p.i. (fever and increased serum IFNγ levels) onward, maintaining drug exposure for 20 days (10 doses at 15 mg/kg every 2 days). Treated animals (G1) exhibited significantly lower rectal temperatures (on days 8 and 9 p.i.), serum IFNγ levels (on day 10 p.i.), and specific IgG levels when compared with non-treated animals (G2). At delivery, significantly higher percentages of healthy lambs were found in infected/treated sheep in G1 (73.3%) and in uninfected sheep in G3 (80%) compared with infected/untreated sheep in G2 (31.3%). Concerning congenital transmission, parasite DNA was neither detected in placenta nor target tissues (brain and lungs) from the fetuses/lambs in G1(infected/treated) and G3 (uninfected). By contrast, parasite DNA was detected in all placentas and lambs from G2 (infected/untreated), except for one sheep that aborted on day 13 p.i.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0144824"},"PeriodicalIF":4.1,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11823607/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142913608","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-13Epub Date: 2024-12-19DOI: 10.1128/aac.01238-24
Lauren Rodriguez, Hery W Lee, Jiani Li, Ross Martin, Dong Han, Simin Xu, Jasmine Moshiri, Nadine Peinovich, Gregory Camus, Jason K Perry, Robert H Hyland, Danielle P Porter, Mazin Abdelghany, Matthias Götte, Charlotte Hedskog
Remdesivir inhibits the SARS-CoV-2 RNA-dependent RNA polymerase (RdRp; Nsp12). Here, we conducted viral resistance analyses from the Phase 3 PINETREE trial of remdesivir in nonhospitalized participants at risk of severe COVID-19. Nasopharyngeal swabs (collected at baseline [Day 1], Days 2, 3, 7, and 14) were eligible for analysis if their viral load was above the lower limit of quantification for the RT-qPCR assay (2228 copies/mL). The SARS-CoV-2 genome was sequenced for all remdesivir participants and 50% of placebo participants (baseline, Days 3, 7, and 14) and for participants who progressed to COVID-19-related hospitalization or all-cause death (all time points). Emergent substitutions in Nsp12 and other replication complex proteins were phenotyped using site-directed mutagenesis in a SARS-CoV-2 subgenomic replicon system. Overall, emergent Nsp12 substitutions were detected in 8/115 (7.0%) remdesivir participants and 7/129 (5.4%) placebo participants (1 substitution overlap between groups). Based on a structural analysis, none of the emergent Nsp12 substitutions were in direct contact with the incoming nucleoside triphosphate substrate, the RNA, or the RNA template 5' overhang. One substitution (A376V) showed reduced susceptibility to remdesivir (12.6-fold change in remdesivir half-maximal concentration [EC50]); it also showed reduced fitness when introduced in the SARS-CoV-2 replicon and virus in vitro. Other substitutions had <1.1-fold change in remdesivir EC50. None of the emergent substitutions in Nsp8, Nsp10, Nsp13, or Nsp14 (remdesivir, 10/115 [8.7%]; placebo, 10/129 [7.8%]) showed reduced remdesivir susceptibility. In conclusion, emergent substitutions in the SARS-CoV-2 RdRp complex with reduced remdesivir susceptibility were uncommon, indicating a high barrier to remdesivir resistance.CLINICAL TRIALSThis study is registered with ClinicalTrials.gov as NCT04501952.
{"title":"SARS-CoV-2 resistance analyses from the Phase 3 PINETREE study of remdesivir treatment in nonhospitalized participants.","authors":"Lauren Rodriguez, Hery W Lee, Jiani Li, Ross Martin, Dong Han, Simin Xu, Jasmine Moshiri, Nadine Peinovich, Gregory Camus, Jason K Perry, Robert H Hyland, Danielle P Porter, Mazin Abdelghany, Matthias Götte, Charlotte Hedskog","doi":"10.1128/aac.01238-24","DOIUrl":"10.1128/aac.01238-24","url":null,"abstract":"<p><p>Remdesivir inhibits the SARS-CoV-2 RNA-dependent RNA polymerase (RdRp; Nsp12). Here, we conducted viral resistance analyses from the Phase 3 PINETREE trial of remdesivir in nonhospitalized participants at risk of severe COVID-19. Nasopharyngeal swabs (collected at baseline [Day 1], Days 2, 3, 7, and 14) were eligible for analysis if their viral load was above the lower limit of quantification for the RT-qPCR assay (2228 copies/mL). The SARS-CoV-2 genome was sequenced for all remdesivir participants and 50% of placebo participants (baseline, Days 3, 7, and 14) and for participants who progressed to COVID-19-related hospitalization or all-cause death (all time points). Emergent substitutions in Nsp12 and other replication complex proteins were phenotyped using site-directed mutagenesis in a SARS-CoV-2 subgenomic replicon system. Overall, emergent Nsp12 substitutions were detected in 8/115 (7.0%) remdesivir participants and 7/129 (5.4%) placebo participants (1 substitution overlap between groups). Based on a structural analysis, none of the emergent Nsp12 substitutions were in direct contact with the incoming nucleoside triphosphate substrate, the RNA, or the RNA template 5' overhang. One substitution (A376V) showed reduced susceptibility to remdesivir (12.6-fold change in remdesivir half-maximal concentration [EC<sub>50</sub>]); it also showed reduced fitness when introduced in the SARS-CoV-2 replicon and virus <i>in vitro</i>. Other substitutions had <1.1-fold change in remdesivir EC<sub>50</sub>. None of the emergent substitutions in Nsp8, Nsp10, Nsp13, or Nsp14 (remdesivir, 10/115 [8.7%]; placebo, 10/129 [7.8%]) showed reduced remdesivir susceptibility. In conclusion, emergent substitutions in the SARS-CoV-2 RdRp complex with reduced remdesivir susceptibility were uncommon, indicating a high barrier to remdesivir resistance.CLINICAL TRIALSThis study is registered with ClinicalTrials.gov as NCT04501952.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0123824"},"PeriodicalIF":4.1,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11823660/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142851760","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-13Epub Date: 2025-01-16DOI: 10.1128/aac.01087-24
Thomas M File, Julio A Ramirez, Ashley M Wilde
Omadacycline is a novel antimicrobial belonging to the tetracycline class. It has the ability to evade both efflux and ribosomal methylation types of resistance and therefore has an expanded spectrum compared to other tetracycline agents. Omadacycline is active against a number of multidrug-resistant bacteria, including macrolide and doxycycline-resistant Streptococcus pneumoniae, methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant Enterococcus, and several enteric gram-negative bacilli. It also has activity against many nontuberculous mycobacterium (NTM) species. It is available both orally and intravenously, which allows for feasible switch therapy. This review will assess the antimicrobial activity, pharmacology, safety, and clinical efficacy of omadacycline and present the opinions of the authors on where to position omadacycline for clinical practice.
{"title":"New Perspectives on Antimicrobial Agents: Omadacycline for community-acquired pneumonia, skin and soft tissue infections, and nontuberculous mycobacteria (focus on <i>M. abscessus</i>).","authors":"Thomas M File, Julio A Ramirez, Ashley M Wilde","doi":"10.1128/aac.01087-24","DOIUrl":"10.1128/aac.01087-24","url":null,"abstract":"<p><p>Omadacycline is a novel antimicrobial belonging to the tetracycline class. It has the ability to evade both efflux and ribosomal methylation types of resistance and therefore has an expanded spectrum compared to other tetracycline agents. Omadacycline is active against a number of multidrug-resistant bacteria, including macrolide and doxycycline-resistant <i>Streptococcus pneumoniae,</i> methicillin-resistant <i>Staphylococcus aureus</i> (MRSA), vancomycin-resistant Enterococcus, and several enteric gram-negative bacilli. It also has activity against many nontuberculous mycobacterium (NTM) species. It is available both orally and intravenously, which allows for feasible switch therapy. This review will assess the antimicrobial activity, pharmacology, safety, and clinical efficacy of omadacycline and present the opinions of the authors on where to position omadacycline for clinical practice.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0108724"},"PeriodicalIF":4.1,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11823662/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142999071","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Brucellosis has therapeutic challenges due to 3%-15% relapses/therapeutic failures (R/TF) after antibiotic treatment. Therefore, determining the antibiotic concentration in tissues, the physiopathological parameters, and the R/TF after treatment is relevant. After exploring different antibiotic quantities, we found that a combined dose of 100 µg/g of doxycycline (for 45 days) and 7.5 µg/g of streptomycin (for 14 days), respectively, achieved therapeutic levels of more than fourfold minimum inhibitory concentrations (MICs) against Brucella abortus in the spleen, liver, bone marrow, and plasma of mice, causing minimal pathophysiological effects. After 30 days of infection, mice received antibiotics, and hematological, histopathological, biochemical, and immunological analyses were performed. After antibiotic therapy, the pathological, hematological, immunological, and physiological profiles paralleled those described in human brucellosis. Treatment lowered antibody titers, reduced proinflammatory cytokines, and reduced inflammation in the target organs for a protracted period. No bacteria were detected in tissues 8 weeks after treatment, suggesting complete recovery. However, despite high doxycycline and streptomycin concentrations in tissues, relapses appeared in 100% of the animals after 182 days post-infection, estimated by the bacterial counts and PCR from organs. This proportion contrasts with the 15% R/TF observed in humans after antibiotic treatments. None of the B. abortus isolated from relapses showed augmented MICs or mutations coding for antibiotic resistance in chromosomal-relevant regions. We discuss whether our findings constitute a general phenomenon or differences in the exhaustive screening method for bacteria detection related to the murine model. Along these lines, we envision likely mechanisms of bacterial persistence in tissues after antibiotic treatment.
{"title":"Reactivation of hidden-latent <i>Brucella</i> infection after doxycycline and streptomycin treatment in mice.","authors":"Eugenia Sancho-Sánchez, Kimberly García-Arteaga, Fabio Granados-Chinchilla, Graciela Artavia, Alejandro Alfaro-Alarcón, Andrés Villalobos-Villalobos, Laura Bouza-Mora, Marcela Suárez-Esquivel, Carlos Chacón-Díaz, Caterina Guzmán-Verri, Edgardo Moreno, Elías Barquero-Calvo","doi":"10.1128/aac.01302-24","DOIUrl":"10.1128/aac.01302-24","url":null,"abstract":"<p><p>Brucellosis has therapeutic challenges due to 3%-15% relapses/therapeutic failures (R/TF) after antibiotic treatment. Therefore, determining the antibiotic concentration in tissues, the physiopathological parameters, and the R/TF after treatment is relevant. After exploring different antibiotic quantities, we found that a combined dose of 100 µg/g of doxycycline (for 45 days) and 7.5 µg/g of streptomycin (for 14 days), respectively, achieved therapeutic levels of more than fourfold minimum inhibitory concentrations (MICs) against <i>Brucella abortus</i> in the spleen, liver, bone marrow, and plasma of mice, causing minimal pathophysiological effects. After 30 days of infection, mice received antibiotics, and hematological, histopathological, biochemical, and immunological analyses were performed. After antibiotic therapy, the pathological, hematological, immunological, and physiological profiles paralleled those described in human brucellosis. Treatment lowered antibody titers, reduced proinflammatory cytokines, and reduced inflammation in the target organs for a protracted period. No bacteria were detected in tissues 8 weeks after treatment, suggesting complete recovery. However, despite high doxycycline and streptomycin concentrations in tissues, relapses appeared in 100% of the animals after 182 days post-infection, estimated by the bacterial counts and PCR from organs. This proportion contrasts with the 15% R/TF observed in humans after antibiotic treatments. None of the <i>B. abortus</i> isolated from relapses showed augmented MICs or mutations coding for antibiotic resistance in chromosomal-relevant regions. We discuss whether our findings constitute a general phenomenon or differences in the exhaustive screening method for bacteria detection related to the murine model. Along these lines, we envision likely mechanisms of bacterial persistence in tissues after antibiotic treatment.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0130224"},"PeriodicalIF":4.1,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11823614/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142913605","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-13Epub Date: 2025-01-14DOI: 10.1128/aac.01575-24
Snježana Mikuličić, Annika Massenberg, Tatjana Döring, Klaus Brandenburg, Thorsten Lang, Luise Florin
Peptide-based therapeutics are gaining attention for their potential to target various viral and host cell factors. One notable example is Pep19-2.5 (Aspidasept), a synthetic anti-lipopolysaccharide peptide that binds to heparan sulfate proteoglycans (HSPGs) and has demonstrated inhibitory effects against certain bacteria and enveloped viruses. This study explores, for the first time, the effectiveness of Pep19-2.5 against a non-enveloped virus, using pseudoviruses of the oncogenic human papillomavirus type 16 (HPV16) as a model. HPV16 infects epithelial cells of the skin and mucosa by using multiple cell surface receptors with initial attachment to HSPGs. Pharmacological inhibition with Pep19-2.5 in HeLa and HaCaT cells resulted in a concentration-dependent reduction of HPV16 PsV infection, with near-complete blockade observed at higher concentrations. The half-maximal inhibitory concentration (IC50) was determined to be 116 nM in HeLa cells and 183 nM in HaCaT cells, highlighting its potent antiviral activity. Our results demonstrate that Pep19-2.5 not only inhibits HPV16 PsV binding to the cell surface but also significantly reduces infection when administered post-binding. Imaging analyses revealed Pep19-2.5-dependent release of large cell-associated crowds of viral particles, suggesting interference with the transfer to secondary receptor molecules. This was corroborated by the effectiveness of Pep19-2.5 in an HSPG-negative cell line, indicating that the peptide disrupts virus binding to both primary and secondary interaction partners. Based on these findings, we propose that the antimicrobial effect of Pep19-2.5 is not limited to HSPG-dependent infections. Additionally, Pep19-2.5 may be a valuable tool for dissecting specific steps in the viral entry process.
{"title":"HSPG-binding peptide Pep19-2.5 is a potent inhibitor of HPV16 infection.","authors":"Snježana Mikuličić, Annika Massenberg, Tatjana Döring, Klaus Brandenburg, Thorsten Lang, Luise Florin","doi":"10.1128/aac.01575-24","DOIUrl":"10.1128/aac.01575-24","url":null,"abstract":"<p><p>Peptide-based therapeutics are gaining attention for their potential to target various viral and host cell factors. One notable example is Pep19-2.5 (Aspidasept), a synthetic anti-lipopolysaccharide peptide that binds to heparan sulfate proteoglycans (HSPGs) and has demonstrated inhibitory effects against certain bacteria and enveloped viruses. This study explores, for the first time, the effectiveness of Pep19-2.5 against a non-enveloped virus, using pseudoviruses of the oncogenic human papillomavirus type 16 (HPV16) as a model. HPV16 infects epithelial cells of the skin and mucosa by using multiple cell surface receptors with initial attachment to HSPGs. Pharmacological inhibition with Pep19-2.5 in HeLa and HaCaT cells resulted in a concentration-dependent reduction of HPV16 PsV infection, with near-complete blockade observed at higher concentrations. The half-maximal inhibitory concentration (IC<sub>50</sub>) was determined to be 116 nM in HeLa cells and 183 nM in HaCaT cells, highlighting its potent antiviral activity. Our results demonstrate that Pep19-2.5 not only inhibits HPV16 PsV binding to the cell surface but also significantly reduces infection when administered post-binding. Imaging analyses revealed Pep19-2.5-dependent release of large cell-associated crowds of viral particles, suggesting interference with the transfer to secondary receptor molecules. This was corroborated by the effectiveness of Pep19-2.5 in an HSPG-negative cell line, indicating that the peptide disrupts virus binding to both primary and secondary interaction partners. Based on these findings, we propose that the antimicrobial effect of Pep19-2.5 is not limited to HSPG-dependent infections. Additionally, Pep19-2.5 may be a valuable tool for dissecting specific steps in the viral entry process.</p>","PeriodicalId":8152,"journal":{"name":"Antimicrobial Agents and Chemotherapy","volume":" ","pages":"e0157524"},"PeriodicalIF":4.1,"publicationDate":"2025-02-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11823620/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142977273","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}