Pub Date : 2026-01-12DOI: 10.1007/s40262-025-01610-4
Yang Wang, Laura H Bukkems, Rob Ter Heine, J G C van Hasselt, S L W Koolen, J J M A Hendrikx, Tom Van der Hulle, Ellen Kapiteijn, Juliette Zwaveling, Annemarie Becker, Michel M van den Heuvel, Willemijn S M E Theelen, Thijs H Oude Munnink, Egbert F Smit, Henk-Jan Guchelaar, Dirk Jan A R Moes
Background: The increasing use of the immune checkpoint inhibitor nivolumab places a significant financial burden on healthcare systems, contributes to environmental concerns, and strains hospital capacities. The nivolumab average exposure and exposure variation of a fixed subcutaneous (SC) dosing regimen (1200 mg every 4 weeks) is significantly higher compared with 3-mg/kg every 2 weeks intravenous dosing.
Objectives: We aimed to develop alternative dosing regimens for SC nivolumab to reduce drug expenses and lower the treatment burden for patients while ensuring effective exposure.
Methods: Population pharmacokinetic simulation was conducted using a population pharmacokinetic model developed by the license holder to explore alternative SC regimens. In this process, patients were divided into three weight groups: less than 60 kg, 60-90 kg, and more than 90 kg. Furthermore, two experimental progressive alternative dosing regimens were developed, one based on a minimum effective concentration-driven approach. The second progressive alternative regimen was based on using the 1200-mg SC formulation as an intravenous infusion.
Results: We developed an alternative bodyweight-based regimen consisting of SC 1200 mg every 7 weeks (<60 kg), 1200 mg every 6 weeks (60-90 kg), and 1200 mg every 5 weeks (>90 kg). This new alternative dosing regimen would save an average of €24,345 (35%) per patient per year compared with SC 1200 mg every 4 weeks. The results for the first experimental, progressive, extended-interval dosing regimen for patients with melanoma indicate that 95% of patients exceed a steady-state trough concentration of 2.5 mg/L when administered SC 1200 mg every 10 weeks. This dosing regimen would decrease the yearly cost from €68,870 to €27,548 (60% less) per patient per year. The second experimental progressive regimen using SC 1200 mg as an intravenous administration every 7 weeks leads to a potential saving of €29,516, which is a 43% decrease compared with the SC 1200-mg approved regimen.
Conclusions: The developed dosing regimen with a bodyweight-dependent interval offers a cost-effective and patient-friendly method to optimize SC nivolumab use while ensuring adequate exposure, which can be directly implemented in clinical practice. Moreover, the two experimental progressive proposed regimens provide a rationale for a clinical non-inferiority study in which alternative dose regimens are compared to standard dosing according to the drug label.
{"title":"Model-Based Alternative Dosing Strategies for Subcutaneous Nivolumab to Improve Cost Effectiveness.","authors":"Yang Wang, Laura H Bukkems, Rob Ter Heine, J G C van Hasselt, S L W Koolen, J J M A Hendrikx, Tom Van der Hulle, Ellen Kapiteijn, Juliette Zwaveling, Annemarie Becker, Michel M van den Heuvel, Willemijn S M E Theelen, Thijs H Oude Munnink, Egbert F Smit, Henk-Jan Guchelaar, Dirk Jan A R Moes","doi":"10.1007/s40262-025-01610-4","DOIUrl":"https://doi.org/10.1007/s40262-025-01610-4","url":null,"abstract":"<p><strong>Background: </strong>The increasing use of the immune checkpoint inhibitor nivolumab places a significant financial burden on healthcare systems, contributes to environmental concerns, and strains hospital capacities. The nivolumab average exposure and exposure variation of a fixed subcutaneous (SC) dosing regimen (1200 mg every 4 weeks) is significantly higher compared with 3-mg/kg every 2 weeks intravenous dosing.</p><p><strong>Objectives: </strong>We aimed to develop alternative dosing regimens for SC nivolumab to reduce drug expenses and lower the treatment burden for patients while ensuring effective exposure.</p><p><strong>Methods: </strong>Population pharmacokinetic simulation was conducted using a population pharmacokinetic model developed by the license holder to explore alternative SC regimens. In this process, patients were divided into three weight groups: less than 60 kg, 60-90 kg, and more than 90 kg. Furthermore, two experimental progressive alternative dosing regimens were developed, one based on a minimum effective concentration-driven approach. The second progressive alternative regimen was based on using the 1200-mg SC formulation as an intravenous infusion.</p><p><strong>Results: </strong>We developed an alternative bodyweight-based regimen consisting of SC 1200 mg every 7 weeks (<60 kg), 1200 mg every 6 weeks (60-90 kg), and 1200 mg every 5 weeks (>90 kg). This new alternative dosing regimen would save an average of €24,345 (35%) per patient per year compared with SC 1200 mg every 4 weeks. The results for the first experimental, progressive, extended-interval dosing regimen for patients with melanoma indicate that 95% of patients exceed a steady-state trough concentration of 2.5 mg/L when administered SC 1200 mg every 10 weeks. This dosing regimen would decrease the yearly cost from €68,870 to €27,548 (60% less) per patient per year. The second experimental progressive regimen using SC 1200 mg as an intravenous administration every 7 weeks leads to a potential saving of €29,516, which is a 43% decrease compared with the SC 1200-mg approved regimen.</p><p><strong>Conclusions: </strong>The developed dosing regimen with a bodyweight-dependent interval offers a cost-effective and patient-friendly method to optimize SC nivolumab use while ensuring adequate exposure, which can be directly implemented in clinical practice. Moreover, the two experimental progressive proposed regimens provide a rationale for a clinical non-inferiority study in which alternative dose regimens are compared to standard dosing according to the drug label.</p>","PeriodicalId":10405,"journal":{"name":"Clinical Pharmacokinetics","volume":" ","pages":""},"PeriodicalIF":4.0,"publicationDate":"2026-01-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145951660","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 : 2026-01-06DOI: 10.1007/s40262-025-01613-1
Jaydeep Sinha, Kanecia Zimmerman, Stephen J Balevic, Chi Hornik, William J Muller, Mobeen Rathore, Marisa Meyer, Yaron Finkelstein, Amira Al-Uzri, Arpita Lakhotia, Stuart Goldstein, Jia-Yuh Chen, Ravinder Anand, Daniel Gonzalez
{"title":"Correction: Population Pharmacokinetic Modeling of Oxcarbazepine and Its Active Metabolite 10‑Monohydroxy Derivative to Inform Dosing in Children with Obesity.","authors":"Jaydeep Sinha, Kanecia Zimmerman, Stephen J Balevic, Chi Hornik, William J Muller, Mobeen Rathore, Marisa Meyer, Yaron Finkelstein, Amira Al-Uzri, Arpita Lakhotia, Stuart Goldstein, Jia-Yuh Chen, Ravinder Anand, Daniel Gonzalez","doi":"10.1007/s40262-025-01613-1","DOIUrl":"https://doi.org/10.1007/s40262-025-01613-1","url":null,"abstract":"","PeriodicalId":10405,"journal":{"name":"Clinical Pharmacokinetics","volume":" ","pages":""},"PeriodicalIF":4.0,"publicationDate":"2026-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145910686","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 : 2026-01-05DOI: 10.1007/s40262-025-01598-x
Dan Tang, Xuehu Gao, Min Zhu, Chang Shu, Yanli Dong, Xiaoling Yu, Hong Chen, Xiaojuan Li, Xiaoli Zhang, Daren Cai, Sheng Feng
Background and objective: INS068 is a new soluble, long-acting insulin analog intended to cover basal insulin requirements in patients with type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM). The purpose of this study was to determine the molar dose ratio of INS068 to insulin degludec (IDeg) by comparing the clinical pharmacokinetic (PK) and pharmacodynamic (PD) profiles of INS068 and IDeg. The PD endpoints include glucose infusion rate (GIR) and glycated hemoglobin (HbA1c).
Method: Population pharmacokinetic and pharmacodynamic (PopPK/PD) analysis was performed to characterize INS068 and IDeg PK and PD profiles. Data from 307 subjects across three Phase I studies and one Phase II study were used to establish the population pharmacokinetic (PopPK) model, of which two euglycemic clamp (Phase I) studies were used to establish PopPK-GIR model, and one Phase II study was used to establish PopPK-HbA1c model. Nonlinear mixed-effects modeling was used to investigate the PK and PD relationships of INS068 and IDeg. Model-based simulations were performed to determine the molar dose ratio of INS068 to IDeg.
Results: The PopPK model of INS068 and IDeg was described by a one-compartment model with linear absorption with a lag time and elimination. Significant covariate effects of body weight, population, and treatment (INS068 vs IDeg) were identified for PK parameters of INS068 and IDeg. However, except for body weight and T2DM patients, the other significant covariates (treatment and T1DM patients) had no clinically relevant effects on PK exposures. The relationship between GIR and insulin concentrations in effect compartment was described by a direct response model with sigmoidal Emax drug effect. The relationship between insulin concentration and HbA1c was well described by an indirect response model. The covariate analysis of the PopPK-GIR and PopPK-HbA1c models showed that treatment (INS068 vs IDeg) had no significant impact on the PD parameters. The results of model-based simulation demonstrated that the PK and PD of INS068 and IDeg were comparable.
Conclusion: This analysis supported molar dose ratio of INS068 to IDeg as 1. INS068 had similar potency compared to IDeg, with one unit of INS068 composed of 6 nmol of active ingredient (1 U = 6 nmol).
背景和目的:INS068是一种新的可溶性、长效胰岛素类似物,旨在满足1型糖尿病(T1DM)和2型糖尿病(T2DM)患者的基础胰岛素需求。本研究的目的是通过比较INS068和IDeg的临床药代动力学(PK)和药效学(PD)谱,确定INS068与胰岛素葡糖苷(IDeg)的摩尔剂量比。PD终点包括葡萄糖输注率(GIR)和糖化血红蛋白(HbA1c)。方法:采用群体药代动力学和药效学(PopPK/PD)方法对INS068和IDeg进行PK和PD谱分析。采用3项I期研究和1项II期研究共307名受试者的数据建立群体药代动力学(PopPK)模型,其中2项正糖钳(I期)研究建立PopPK- gir模型,1项II期研究建立PopPK- hba1c模型。采用非线性混合效应模型研究了INS068和IDeg的PK和PD关系。通过模型模拟确定了INS068与IDeg的摩尔剂量比。结果:INS068和IDeg的PopPK模型均为线性吸收的单室模型,存在时滞和消除。体重、种群和处理(INS068 vs IDeg)对INS068和IDeg的PK参数有显著的协变量影响。然而,除了体重和T2DM患者外,其他重要协变量(治疗和T1DM患者)对PK暴露没有临床相关的影响。效应室胰岛素浓度与GIR之间的关系采用具有s型Emax药物效应的直接反应模型。胰岛素浓度与HbA1c之间的关系通过间接反应模型得到了很好的描述。PopPK-GIR和PopPK-HbA1c模型的协变量分析显示,治疗(INS068 vs IDeg)对PD参数没有显著影响。基于模型的模拟结果表明,INS068和IDeg的PK和PD具有可比性。结论:该分析支持INS068与IDeg的摩尔剂量比为1。与IDeg相比,INS068的效价相似,1单位INS068含有6 nmol的有效成分(1 U = 6 nmol)。
{"title":"Population Pharmacokinetic and Pharmacodynamic Modeling to Support Molar Dose Ratio Determination of Long-Acting Insulin Analogs.","authors":"Dan Tang, Xuehu Gao, Min Zhu, Chang Shu, Yanli Dong, Xiaoling Yu, Hong Chen, Xiaojuan Li, Xiaoli Zhang, Daren Cai, Sheng Feng","doi":"10.1007/s40262-025-01598-x","DOIUrl":"https://doi.org/10.1007/s40262-025-01598-x","url":null,"abstract":"<p><strong>Background and objective: </strong>INS068 is a new soluble, long-acting insulin analog intended to cover basal insulin requirements in patients with type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM). The purpose of this study was to determine the molar dose ratio of INS068 to insulin degludec (IDeg) by comparing the clinical pharmacokinetic (PK) and pharmacodynamic (PD) profiles of INS068 and IDeg. The PD endpoints include glucose infusion rate (GIR) and glycated hemoglobin (HbA1c).</p><p><strong>Method: </strong>Population pharmacokinetic and pharmacodynamic (PopPK/PD) analysis was performed to characterize INS068 and IDeg PK and PD profiles. Data from 307 subjects across three Phase I studies and one Phase II study were used to establish the population pharmacokinetic (PopPK) model, of which two euglycemic clamp (Phase I) studies were used to establish PopPK-GIR model, and one Phase II study was used to establish PopPK-HbA1c model. Nonlinear mixed-effects modeling was used to investigate the PK and PD relationships of INS068 and IDeg. Model-based simulations were performed to determine the molar dose ratio of INS068 to IDeg.</p><p><strong>Results: </strong>The PopPK model of INS068 and IDeg was described by a one-compartment model with linear absorption with a lag time and elimination. Significant covariate effects of body weight, population, and treatment (INS068 vs IDeg) were identified for PK parameters of INS068 and IDeg. However, except for body weight and T2DM patients, the other significant covariates (treatment and T1DM patients) had no clinically relevant effects on PK exposures. The relationship between GIR and insulin concentrations in effect compartment was described by a direct response model with sigmoidal E<sub>max</sub> drug effect. The relationship between insulin concentration and HbA1c was well described by an indirect response model. The covariate analysis of the PopPK-GIR and PopPK-HbA1c models showed that treatment (INS068 vs IDeg) had no significant impact on the PD parameters. The results of model-based simulation demonstrated that the PK and PD of INS068 and IDeg were comparable.</p><p><strong>Conclusion: </strong>This analysis supported molar dose ratio of INS068 to IDeg as 1. INS068 had similar potency compared to IDeg, with one unit of INS068 composed of 6 nmol of active ingredient (1 U = 6 nmol).</p>","PeriodicalId":10405,"journal":{"name":"Clinical Pharmacokinetics","volume":" ","pages":""},"PeriodicalIF":4.0,"publicationDate":"2026-01-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145899355","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 : 2026-01-04DOI: 10.1007/s40262-025-01597-y
Gerbert Coen de Waard, Qiaolin Zhao, Ewout S Veltman, Jakob van Oldenrijk, P Koen Bos, Soma Bahmany, Anouk Muller, Tim Preijers, Birgit C P Koch
Background and objective: Prosthetic joint infections are serious infections that require perioperative antibiotic prophylaxis. Cefuroxime may be used as prophylaxis, and this study aims to evaluate the current dosing regimen to prevent infection during the perioperative period by performing population pharmacokinetic (PK) analysis of plasma and bone in patients with prosthetic joint infection (PJI).
Methods: Both plasma and bone samples were taken perioperatively and analyzed using NONMEM. Monte Carlo simulations were performed to determine the probability of target attainment (PTA) in both plasma and bone of various dosing regimens.
Results: A total of 44 plasma and 38 bone samples from 25 patients were included. The median bone concentration at 30-60 min after administration was 18.2 (9.9-25.3) mg/L and the bone-to-plasma ratio was 0.269 (interquartile range [IQR] 0.179-0.269), while the median bone concentration at 90-120 min after administration was 12.6 (5.9-18.6) mg/L and the bone-to-plasma ratio was 0.125 (IQR 0.073-0.160). A two-compartment model with allometric scaling and proportional errors best described plasma and bone concentrations. Only estimated glomerular filtration rate could partly explain the inter-individual variability (IIV) of clearance (CL). A single dose of 1500 mg cefuroxime failed to maintain bone concentrations above target concentrations for Staphylococcus aureus beyond 3.83 h post administration.
Conclusion: A population PK model was developed to characterize the disposition of cefuroxime in both plasma and bone compartments. Although adequate cefuroxime bone penetration within 1 h was observed in patients with PJI with rapid clearance, simulations predicted less optimal levels of cefuroxime after 3.5 h. The clinical implications need to be researched and confirmed.
{"title":"Population Pharmacokinetics of Intravenous Cefuroxime in Plasma and Bone of Patients with Prosthetic Joint Infection: Is There Room for Dose Optimization?","authors":"Gerbert Coen de Waard, Qiaolin Zhao, Ewout S Veltman, Jakob van Oldenrijk, P Koen Bos, Soma Bahmany, Anouk Muller, Tim Preijers, Birgit C P Koch","doi":"10.1007/s40262-025-01597-y","DOIUrl":"https://doi.org/10.1007/s40262-025-01597-y","url":null,"abstract":"<p><strong>Background and objective: </strong>Prosthetic joint infections are serious infections that require perioperative antibiotic prophylaxis. Cefuroxime may be used as prophylaxis, and this study aims to evaluate the current dosing regimen to prevent infection during the perioperative period by performing population pharmacokinetic (PK) analysis of plasma and bone in patients with prosthetic joint infection (PJI).</p><p><strong>Methods: </strong>Both plasma and bone samples were taken perioperatively and analyzed using NONMEM. Monte Carlo simulations were performed to determine the probability of target attainment (PTA) in both plasma and bone of various dosing regimens.</p><p><strong>Results: </strong>A total of 44 plasma and 38 bone samples from 25 patients were included. The median bone concentration at 30-60 min after administration was 18.2 (9.9-25.3) mg/L and the bone-to-plasma ratio was 0.269 (interquartile range [IQR] 0.179-0.269), while the median bone concentration at 90-120 min after administration was 12.6 (5.9-18.6) mg/L and the bone-to-plasma ratio was 0.125 (IQR 0.073-0.160). A two-compartment model with allometric scaling and proportional errors best described plasma and bone concentrations. Only estimated glomerular filtration rate could partly explain the inter-individual variability (IIV) of clearance (CL). A single dose of 1500 mg cefuroxime failed to maintain bone concentrations above target concentrations for Staphylococcus aureus beyond 3.83 h post administration.</p><p><strong>Conclusion: </strong>A population PK model was developed to characterize the disposition of cefuroxime in both plasma and bone compartments. Although adequate cefuroxime bone penetration within 1 h was observed in patients with PJI with rapid clearance, simulations predicted less optimal levels of cefuroxime after 3.5 h. The clinical implications need to be researched and confirmed.</p>","PeriodicalId":10405,"journal":{"name":"Clinical Pharmacokinetics","volume":" ","pages":""},"PeriodicalIF":4.0,"publicationDate":"2026-01-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145896445","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 : 2026-01-01Epub Date: 2025-11-03DOI: 10.1007/s40262-025-01577-2
Stef Schouwenburg, Tim Preijers, Roelie M Wösten-van Asperen, Stan J F Hartman, Saskia N de Wildt, Matthijs de Hoog, Birgit C P Koch, Alan Abdulla, Enno D Wildschut
Background and objective: Cefuroxime is a widely prescribed beta-lactam antibiotic, particularly in pediatric cardiac and medical-surgical intensive care units. The aim of this study was to describe intravenous cefuroxime disposition in critically ill pediatric patients. Moreover, target attainment of currently applied dosing regimens was evaluated, and suggestions for improving these dosing regimens were provided.
Methods: Two datasets were pooled for population pharmacokinetic (popPK) analysis, using NONMEM version 7.5. To assess the optimal target attainment (> 90% of patients with 100% time [T] > [4×] minimal inhibitory concentration [MIC]8mg/L), pharmacokinetic (PK) profiles of different dosage regimens (intermittent/continuous) were simulated using the estimated popPK parameters.
Results: The cohort consisted of 45 pediatric patients with a median (interquartile range [IQR]) age of 391 days [31-3505] and body weight of 9.0 kg [5.0-29.8]. A two-compartment popPK model with first-order elimination and allometric scaling best described cefuroxime disposition. Intravenous cefuroxime clearance was estimated at 5.29 L/h/70 kg. Postnatal age and creatinine clearance (mL/min/1.73 m2) were the best descriptive covariates for the maturation of cefuroxime clearance. Simulations evaluating the current cefuroxime dosing regimens stratified for estimated glomerular filtration rate (eGFR) levels illustrated moderate (< 90%) (eGFR < 30 and 30-80 mL/min/1.73 m2) and poor (< 20%) (eGFR 80-120 and > 120 mL/min/1.73 m2) cefuroxime target attainment across the entire age range. Alternative dosing regimens, including four times daily schedules and continuous infusion, improved target attainment, particularly in older children and those with augmented renal clearance.
Conclusions: These findings indicate that underexposure due to augmented renal function is possible when applying the current cefuroxime dosing regimens. Future research should focus on individualized dosing strategies to optimize cefuroxime exposure and efficacy in pediatric populations.
{"title":"Low Target Attainment of Intravenous Cefuroxime in Critically Ill Term Neonates and Children: A Pooled Population Pharmacokinetics Study.","authors":"Stef Schouwenburg, Tim Preijers, Roelie M Wösten-van Asperen, Stan J F Hartman, Saskia N de Wildt, Matthijs de Hoog, Birgit C P Koch, Alan Abdulla, Enno D Wildschut","doi":"10.1007/s40262-025-01577-2","DOIUrl":"10.1007/s40262-025-01577-2","url":null,"abstract":"<p><strong>Background and objective: </strong>Cefuroxime is a widely prescribed beta-lactam antibiotic, particularly in pediatric cardiac and medical-surgical intensive care units. The aim of this study was to describe intravenous cefuroxime disposition in critically ill pediatric patients. Moreover, target attainment of currently applied dosing regimens was evaluated, and suggestions for improving these dosing regimens were provided.</p><p><strong>Methods: </strong>Two datasets were pooled for population pharmacokinetic (popPK) analysis, using NONMEM version 7.5. To assess the optimal target attainment (> 90% of patients with 100% time [T] > [4×] minimal inhibitory concentration [MIC]<sub>8mg/L</sub>), pharmacokinetic (PK) profiles of different dosage regimens (intermittent/continuous) were simulated using the estimated popPK parameters.</p><p><strong>Results: </strong>The cohort consisted of 45 pediatric patients with a median (interquartile range [IQR]) age of 391 days [31-3505] and body weight of 9.0 kg [5.0-29.8]. A two-compartment popPK model with first-order elimination and allometric scaling best described cefuroxime disposition. Intravenous cefuroxime clearance was estimated at 5.29 L/h/70 kg. Postnatal age and creatinine clearance (mL/min/1.73 m<sup>2</sup>) were the best descriptive covariates for the maturation of cefuroxime clearance. Simulations evaluating the current cefuroxime dosing regimens stratified for estimated glomerular filtration rate (eGFR) levels illustrated moderate (< 90%) (eGFR < 30 and 30-80 mL/min/1.73 m<sup>2</sup>) and poor (< 20%) (eGFR 80-120 and > 120 mL/min/1.73 m<sup>2</sup>) cefuroxime target attainment across the entire age range. Alternative dosing regimens, including four times daily schedules and continuous infusion, improved target attainment, particularly in older children and those with augmented renal clearance.</p><p><strong>Conclusions: </strong>These findings indicate that underexposure due to augmented renal function is possible when applying the current cefuroxime dosing regimens. Future research should focus on individualized dosing strategies to optimize cefuroxime exposure and efficacy in pediatric populations.</p>","PeriodicalId":10405,"journal":{"name":"Clinical Pharmacokinetics","volume":" ","pages":"97-108"},"PeriodicalIF":4.0,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12783212/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145437136","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 : 2026-01-01Epub Date: 2025-10-18DOI: 10.1007/s40262-025-01578-1
Valentin Al Jalali, Martin Bauer, Anselm Jorda, Felix Bergmann, Michael Wölfl-Duchek, Richard Partl, Brigitta Vcelar, Dietmar Katinger, Rawad Bashur, Harald Schnidar, Markus Zeitlinger
Background: Excessive production of reactive oxygen species (ROS), particularly superoxide anion ( ), is a key mechanism in diseases such as cancer, inflammatory disorders, neurodegenerative conditions, and metabolic diseases. Evidence also suggests that microgravity-induced oxidative stress, primarily driven by elevated levels, may contribute to the adverse physiological effects observed in astronauts during extended space missions. Superoxide dismutase (SOD) is critical for mitigating oxidative stress, and exogenous SOD supplementation offers a potential therapeutic strategy.
Methods: This randomized, double-blind, placebo-controlled Phase I study evaluated the safety, tolerability, and pharmacokinetics of recombinant human Cu/Zn-SOD (rhSOD, SOD1) following subcutaneous administration of 40 mg every 12 hours in 16 healthy volunteers. Eight subjects were enrolled each in the single-dose (SD) and multiple-dose (MD) cohorts. Study assessments, including pharmacokinetic sampling, were performed for 72 (SD) or 92 hours (MD). Injection site erythema was objectively assessed using the innovative Standardized Erythema Value (SEV*) method, derived from photographs taken with Scarletred®Vision software (SCARLETRED Holding GmbH).
Results: No serious adverse events occurred, and all treatment-related adverse events were mild. Injection site erythema was objectively assessed using the innovative standardized erythema value (SEV*) method, derived from photographs taken with Scarletred® Vision software (SCARLETRED Holding GmbH). The Visual Analog Scale scores and SEV* assessments were comparable between the rhSOD and placebo groups. Beyond safety and tolerability, pharmacokinetic analysis revealed that the volume of distribution, clearance, and half-life at presumed steady state were 129 ± 66.3 L, 5.97 ± 1.25 L/h, and 15.0 ± 6.69 h, respectively. Compared with the rapid systemic elimination after intravenous administration of SOD, subcutaneous administration resulted in a favorable plasma concentration-time profile.
Conclusions: These findings suggest that subcutaneous rhSOD may be a promising therapeutic candidate for conditions characterized by excessive exposure or diminished endogenous SOD activity. Further clinical studies are warranted to assess its anti-inflammatory potential in relevant patient populations. EudraCT Number: 2022-000173-11.
{"title":"Randomized, Double-Blind, Phase I Pharmacokinetic Study of Subcutaneous Recombinant Human Superoxide Dismutase (rhSOD) in Healthy Volunteers.","authors":"Valentin Al Jalali, Martin Bauer, Anselm Jorda, Felix Bergmann, Michael Wölfl-Duchek, Richard Partl, Brigitta Vcelar, Dietmar Katinger, Rawad Bashur, Harald Schnidar, Markus Zeitlinger","doi":"10.1007/s40262-025-01578-1","DOIUrl":"10.1007/s40262-025-01578-1","url":null,"abstract":"<p><strong>Background: </strong>Excessive production of reactive oxygen species (ROS), particularly superoxide anion ( <math><msubsup><mtext>O</mtext> <mrow><mn>2</mn></mrow> <mrow><mo>·</mo> <mo>-</mo></mrow> </msubsup> </math> ), is a key mechanism in diseases such as cancer, inflammatory disorders, neurodegenerative conditions, and metabolic diseases. Evidence also suggests that microgravity-induced oxidative stress, primarily driven by elevated <math><msubsup><mtext>O</mtext> <mrow><mn>2</mn></mrow> <mrow><mo>·</mo> <mo>-</mo></mrow> </msubsup> </math> levels, may contribute to the adverse physiological effects observed in astronauts during extended space missions. Superoxide dismutase (SOD) is critical for mitigating oxidative stress, and exogenous SOD supplementation offers a potential therapeutic strategy.</p><p><strong>Methods: </strong>This randomized, double-blind, placebo-controlled Phase I study evaluated the safety, tolerability, and pharmacokinetics of recombinant human Cu/Zn-SOD (rhSOD, SOD1) following subcutaneous administration of 40 mg every 12 hours in 16 healthy volunteers. Eight subjects were enrolled each in the single-dose (SD) and multiple-dose (MD) cohorts. Study assessments, including pharmacokinetic sampling, were performed for 72 (SD) or 92 hours (MD). Injection site erythema was objectively assessed using the innovative Standardized Erythema Value (SEV*) method, derived from photographs taken with Scarletred®Vision software (SCARLETRED Holding GmbH).</p><p><strong>Results: </strong>No serious adverse events occurred, and all treatment-related adverse events were mild. Injection site erythema was objectively assessed using the innovative standardized erythema value (SEV*) method, derived from photographs taken with Scarletred<sup>®</sup> Vision software (SCARLETRED Holding GmbH). The Visual Analog Scale scores and SEV* assessments were comparable between the rhSOD and placebo groups. Beyond safety and tolerability, pharmacokinetic analysis revealed that the volume of distribution, clearance, and half-life at presumed steady state were 129 ± 66.3 L, 5.97 ± 1.25 L/h, and 15.0 ± 6.69 h, respectively. Compared with the rapid systemic elimination after intravenous administration of SOD, subcutaneous administration resulted in a favorable plasma concentration-time profile.</p><p><strong>Conclusions: </strong>These findings suggest that subcutaneous rhSOD may be a promising therapeutic candidate for conditions characterized by excessive <math><msubsup><mtext>O</mtext> <mrow><mn>2</mn></mrow> <mrow><mo>·</mo> <mo>-</mo></mrow> </msubsup> </math> exposure or diminished endogenous SOD activity. Further clinical studies are warranted to assess its anti-inflammatory potential in relevant patient populations. EudraCT Number: 2022-000173-11.</p>","PeriodicalId":10405,"journal":{"name":"Clinical Pharmacokinetics","volume":" ","pages":"71-80"},"PeriodicalIF":4.0,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12783157/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145318070","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 : 2026-01-01Epub Date: 2025-12-05DOI: 10.1007/s40262-025-01603-3
Arnaud De Clercq, Jeroen Vervalcke, Cristina De Petter, Stef De Ryck, Tania Desmet, Peter De Paepe, Mirko Petrovic, Pieter A De Cock
Background and objective: Fluoroquinolones are among the most frequently prescribed antibiotics in older adults. Age-related pharmacokinetic (PK) changes can affect treatment efficacy and increase the risk of adverse drug reactions. This systematic review provides a comprehensive overview of the current knowledge on the PK and PK/pharmacodynamic (PD) target attainment of fluoroquinolones in older adults to inform strategies for personalised fluoroquinolone therapy.
Methods: A comprehensive search of the Medline, Embase, Web of Science and Scopus databases was conducted. Relevant articles published before 1 December 2024 were included when they contained data on the PK of fluoroquinolones in older adults (median age ≥ 65 years). Extracted information included PK parameters, significant covariates influencing PK parameters, PK/PD target attainment rates and dosing recommendations. The ClinPK Statement checklist was used for quality grading.
Results: Fifty-five articles were included in this review, encompassing a total of 1542 non-critically ill older adults and 585 younger controls. Eight articles (14.5%) identified covariates with a significant effect on PK parameters. Most of these covariates (66.7%) were indicators of renal function. PK/PD target attainment was assessed in 30.9%, and dosing recommendations were provided in 61.8% of all included PK studies. Studies had an average quality score of 65.9% (standard deviation, SD ± 12.3%).
Conclusions: High-quality PK studies on fluoroquinolones in older adults remain sparse. While a substantial amount of the included articles provided dosing recommendations, only a minority did so on the basis of PK/PD target attainment data. The limited application of advanced PK/PD modeling and simulation approaches hampers the development of evidence-based, individualised fluoroquinolone dosing strategies in older adults.
Trial registration: Prospectively registered in PROSPERO.
背景和目的:氟喹诺酮类药物是老年人最常用的抗生素之一。与年龄相关的药代动力学(PK)变化会影响治疗效果,增加药物不良反应的风险。本系统综述全面概述了目前关于老年人氟喹诺酮类药物的PK和PK/药效学(PD)目标实现的知识,为个性化氟喹诺酮类药物治疗策略提供信息。方法:综合检索Medline、Embase、Web of Science和Scopus数据库。纳入2024年12月1日之前发表的相关文章,包括老年人(中位年龄≥65岁)氟喹诺酮类药物的PK数据。提取的信息包括PK参数、影响PK参数的显著协变量、PK/PD目标完成率和剂量建议。使用ClinPK声明检查表进行质量分级。结果:本综述纳入55篇文章,包括1542名非危重老年人和585名年轻对照。8篇(14.5%)文章确定了对PK参数有显著影响的协变量。大多数协变量(66.7%)为肾功能指标。在所有纳入的PK研究中,有30.9%评估了PK/PD目标的实现,61.8%提供了剂量建议。研究的平均质量评分为65.9%(标准差,SD±12.3%)。结论:老年人氟喹诺酮类药物的高质量PK研究仍然很少。虽然大量纳入的文章提供了剂量建议,但只有少数是基于PK/PD目标实现数据。先进的PK/PD建模和模拟方法的有限应用阻碍了以证据为基础的老年人个体化氟喹诺酮给药策略的发展。试验注册:预期在PROSPERO注册。试验注册号:CRD42023480126。报名日期:2023年11月17日。
{"title":"Pharmacokinetics and Target Attainment of Fluoroquinolones in Older Adults: A Systematic Review.","authors":"Arnaud De Clercq, Jeroen Vervalcke, Cristina De Petter, Stef De Ryck, Tania Desmet, Peter De Paepe, Mirko Petrovic, Pieter A De Cock","doi":"10.1007/s40262-025-01603-3","DOIUrl":"10.1007/s40262-025-01603-3","url":null,"abstract":"<p><strong>Background and objective: </strong>Fluoroquinolones are among the most frequently prescribed antibiotics in older adults. Age-related pharmacokinetic (PK) changes can affect treatment efficacy and increase the risk of adverse drug reactions. This systematic review provides a comprehensive overview of the current knowledge on the PK and PK/pharmacodynamic (PD) target attainment of fluoroquinolones in older adults to inform strategies for personalised fluoroquinolone therapy.</p><p><strong>Methods: </strong>A comprehensive search of the Medline, Embase, Web of Science and Scopus databases was conducted. Relevant articles published before 1 December 2024 were included when they contained data on the PK of fluoroquinolones in older adults (median age ≥ 65 years). Extracted information included PK parameters, significant covariates influencing PK parameters, PK/PD target attainment rates and dosing recommendations. The ClinPK Statement checklist was used for quality grading.</p><p><strong>Results: </strong>Fifty-five articles were included in this review, encompassing a total of 1542 non-critically ill older adults and 585 younger controls. Eight articles (14.5%) identified covariates with a significant effect on PK parameters. Most of these covariates (66.7%) were indicators of renal function. PK/PD target attainment was assessed in 30.9%, and dosing recommendations were provided in 61.8% of all included PK studies. Studies had an average quality score of 65.9% (standard deviation, SD ± 12.3%).</p><p><strong>Conclusions: </strong>High-quality PK studies on fluoroquinolones in older adults remain sparse. While a substantial amount of the included articles provided dosing recommendations, only a minority did so on the basis of PK/PD target attainment data. The limited application of advanced PK/PD modeling and simulation approaches hampers the development of evidence-based, individualised fluoroquinolone dosing strategies in older adults.</p><p><strong>Trial registration: </strong>Prospectively registered in PROSPERO.</p><p><strong>Trial registration number: </strong>CRD42023480126. Registration date: 17/11/2023.</p>","PeriodicalId":10405,"journal":{"name":"Clinical Pharmacokinetics","volume":" ","pages":"27-69"},"PeriodicalIF":4.0,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145676286","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 : 2026-01-01DOI: 10.1007/s40262-025-01600-6
Tessa Rademaker, Yeonkyung Goo, May Ching Soh, Dharmintra Pasupathy, Chong Wong, Andrew Bleasel, Jan-Willem Alffenaar
{"title":"Correction: How is Levetiracetam Monotherapy Currently Monitored in Pregnancy? A Systematic Review.","authors":"Tessa Rademaker, Yeonkyung Goo, May Ching Soh, Dharmintra Pasupathy, Chong Wong, Andrew Bleasel, Jan-Willem Alffenaar","doi":"10.1007/s40262-025-01600-6","DOIUrl":"10.1007/s40262-025-01600-6","url":null,"abstract":"","PeriodicalId":10405,"journal":{"name":"Clinical Pharmacokinetics","volume":" ","pages":"165"},"PeriodicalIF":4.0,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12783147/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145647529","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 : 2026-01-01Epub Date: 2025-10-21DOI: 10.1007/s40262-025-01580-7
Tessa Rademaker, Yeonkyung Goo, May Ching Soh, Dharmintra Pasupathy, Chong Wong, Andrew Bleasel, Jan-Willem Alffenaar
Background: Levetiracetam is commonly used as antiepileptic drug during pregnancy and has a well-established safety profile in that setting. Pregnancy-related pharmacokinetic changes may result in decreased levetiracetam levels and increased seizure frequency during pregnancy. Therapeutic drug monitoring (TDM) of levetiracetam levels may facilitate dose adjustment. Although levetiracetam TDM is widely used in pregnancy, robust guidelines are still lacking, resulting in heterogeneous use of TDM.
Objectives: The aim of this review was to provide insight into levetiracetam TDM strategies utilised during pregnancy.
Methods: A systematic search up to 17 April 2025 was carried out using MEDLINE, Embase, APA PsycINFO, and Cochrane Central Register of Controlled Trials through Ovid. Studies were eligible for inclusion if data on levetiracetam dosing, concentrations, monitoring, efficacy, or safety during pregnancy were available. All articles were assessed for the risk of bias, and relevant data were extracted.
Results: The ten studies included revealed significant variability in epilepsy monitoring during pregnancy, TDM strategies, and dose adjustments. An increase in seizure frequency during pregnancy was described. However, data on levetiracetam maternal and foetal safety were limited.
Conclusions: This review highlights the heterogeneity in levetiracetam TDM strategies in pregnancy. The pharmacokinetic changes during pregnancy require dose adjustments to maintain seizure control, but no standardised TDM protocol is available. Future research should focus on standardizing TDM strategies, validating target concentration thresholds, and assessing long-term maternal and foetal safety.
背景:左乙拉西坦是妊娠期常用的抗癫痫药物,在妊娠期具有良好的安全性。妊娠相关药代动力学改变可导致妊娠期间左乙拉西坦水平降低和癫痫发作频率增加。治疗药物监测(TDM)水平的左乙拉西坦可能有助于剂量调整。虽然左乙拉西坦TDM在妊娠期被广泛使用,但缺乏强有力的指南,导致TDM的使用不均匀。目的:本综述的目的是为妊娠期间使用的左乙拉西坦TDM策略提供见解。方法:通过Ovid使用MEDLINE、Embase、APA PsycINFO和Cochrane Central Register of Controlled Trials进行截至2025年4月17日的系统检索。如果有关于妊娠期间左乙拉西坦剂量、浓度、监测、疗效或安全性的数据,研究才有资格纳入。对所有文章进行偏倚风险评估,并提取相关数据。结果:这10项研究揭示了妊娠期间癫痫监测、TDM策略和剂量调整的显著差异。怀孕期间癫痫发作频率增加。然而,关于左乙拉西坦母婴安全性的数据有限。结论:本综述强调了妊娠期左乙拉西坦TDM策略的异质性。妊娠期间的药代动力学变化需要调整剂量以维持癫痫的控制,但目前尚无标准化的TDM方案。未来的研究应着眼于规范TDM策略、验证靶浓度阈值以及评估母婴长期安全性。
{"title":"How is Levetiracetam Monotherapy Currently Monitored in Pregnancy? A Systematic Review.","authors":"Tessa Rademaker, Yeonkyung Goo, May Ching Soh, Dharmintra Pasupathy, Chong Wong, Andrew Bleasel, Jan-Willem Alffenaar","doi":"10.1007/s40262-025-01580-7","DOIUrl":"10.1007/s40262-025-01580-7","url":null,"abstract":"<p><strong>Background: </strong>Levetiracetam is commonly used as antiepileptic drug during pregnancy and has a well-established safety profile in that setting. Pregnancy-related pharmacokinetic changes may result in decreased levetiracetam levels and increased seizure frequency during pregnancy. Therapeutic drug monitoring (TDM) of levetiracetam levels may facilitate dose adjustment. Although levetiracetam TDM is widely used in pregnancy, robust guidelines are still lacking, resulting in heterogeneous use of TDM.</p><p><strong>Objectives: </strong>The aim of this review was to provide insight into levetiracetam TDM strategies utilised during pregnancy.</p><p><strong>Methods: </strong>A systematic search up to 17 April 2025 was carried out using MEDLINE, Embase, APA PsycINFO, and Cochrane Central Register of Controlled Trials through Ovid. Studies were eligible for inclusion if data on levetiracetam dosing, concentrations, monitoring, efficacy, or safety during pregnancy were available. All articles were assessed for the risk of bias, and relevant data were extracted.</p><p><strong>Results: </strong>The ten studies included revealed significant variability in epilepsy monitoring during pregnancy, TDM strategies, and dose adjustments. An increase in seizure frequency during pregnancy was described. However, data on levetiracetam maternal and foetal safety were limited.</p><p><strong>Conclusions: </strong>This review highlights the heterogeneity in levetiracetam TDM strategies in pregnancy. The pharmacokinetic changes during pregnancy require dose adjustments to maintain seizure control, but no standardised TDM protocol is available. Future research should focus on standardizing TDM strategies, validating target concentration thresholds, and assessing long-term maternal and foetal safety.</p>","PeriodicalId":10405,"journal":{"name":"Clinical Pharmacokinetics","volume":" ","pages":"11-25"},"PeriodicalIF":4.0,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12783189/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145343826","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 : 2026-01-01Epub Date: 2025-11-03DOI: 10.1007/s40262-025-01586-1
Pier Giorgio Cojutti, Pasquale Maria Berrino, Valeria Rotaru, Lorenzo Bianchi, Riccardo Schiavina, Eugenio Brunocilla, Pierluigi Viale, Federico Pea
Background and objective: Fosfomycin trometamol may be a valuable option for antimicrobial prophylaxis before urological surgery for benign prostatic hyperplasia. The objective is to develop a population pharmacokinetic model of a novel oral fosfomycin prophylactic scheme in the plasma and prostate of patients undergoing endoscopic surgery for benign prostatic hyperplasia.
Methods: One- and two-compartment plasma pharmacokinetic models were fitted to fosfomycin plasma data, and different plasma-prostate linked models were tested by means of non-linear mixed effects modelling. Monte Carlo simulation was used to obtain 1000-subject concentration-time profiles of fosfomycin in the prostate. Probabilities of target attainment ≥ 90% of an area under the plasma concentration-time curve/minimum inhibitory concentration (MIC) > 83.3 and of a 70%t > MIC in the prostate were considered as optimal. Cumulative fractions of response against both wild-type and extended-spectrum beta-lactamase-producing Escherichia coli were calculated.
Results: A total of 104 patients, each providing a pair of plasma and prostate concomitant samples were included in the study. A one-compartment pharmacokinetic model was used to describe plasma fosfomycin concentration. Fosfomycin plasma-prostate relationships were adequately described by a direct response model with a power function. Simulations showed that fosfomycin disposition in the prostate was closely related to that in plasma. Optimal probabilities of target attainments were ensured against Enterobacterales having an MIC up to 0.5-1 mg/L in the 12 h vulnerable period after completing the prophylactic scheme.
Conclusion: A prophylactic regimen of two doses of oral fosfomycin trometamol 3 g 12 h apart before undergoing prostatic surgery may grant effective concentrations in the prostatic tissue of patients for a 12 h vulnerable period.
{"title":"Population Pharmacokinetics of a Novel Oral Fosfomycin Prophylactic Scheme in the Plasma and Prostate of Patients Undergoing Endoscopic Surgery for Benign Prostatic Hyperplasia.","authors":"Pier Giorgio Cojutti, Pasquale Maria Berrino, Valeria Rotaru, Lorenzo Bianchi, Riccardo Schiavina, Eugenio Brunocilla, Pierluigi Viale, Federico Pea","doi":"10.1007/s40262-025-01586-1","DOIUrl":"10.1007/s40262-025-01586-1","url":null,"abstract":"<p><strong>Background and objective: </strong>Fosfomycin trometamol may be a valuable option for antimicrobial prophylaxis before urological surgery for benign prostatic hyperplasia. The objective is to develop a population pharmacokinetic model of a novel oral fosfomycin prophylactic scheme in the plasma and prostate of patients undergoing endoscopic surgery for benign prostatic hyperplasia.</p><p><strong>Methods: </strong>One- and two-compartment plasma pharmacokinetic models were fitted to fosfomycin plasma data, and different plasma-prostate linked models were tested by means of non-linear mixed effects modelling. Monte Carlo simulation was used to obtain 1000-subject concentration-time profiles of fosfomycin in the prostate. Probabilities of target attainment ≥ 90% of an area under the plasma concentration-time curve/minimum inhibitory concentration (MIC) > 83.3 and of a 70%t > MIC in the prostate were considered as optimal. Cumulative fractions of response against both wild-type and extended-spectrum beta-lactamase-producing Escherichia coli were calculated.</p><p><strong>Results: </strong>A total of 104 patients, each providing a pair of plasma and prostate concomitant samples were included in the study. A one-compartment pharmacokinetic model was used to describe plasma fosfomycin concentration. Fosfomycin plasma-prostate relationships were adequately described by a direct response model with a power function. Simulations showed that fosfomycin disposition in the prostate was closely related to that in plasma. Optimal probabilities of target attainments were ensured against Enterobacterales having an MIC up to 0.5-1 mg/L in the 12 h vulnerable period after completing the prophylactic scheme.</p><p><strong>Conclusion: </strong>A prophylactic regimen of two doses of oral fosfomycin trometamol 3 g 12 h apart before undergoing prostatic surgery may grant effective concentrations in the prostatic tissue of patients for a 12 h vulnerable period.</p>","PeriodicalId":10405,"journal":{"name":"Clinical Pharmacokinetics","volume":" ","pages":"109-118"},"PeriodicalIF":4.0,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12783171/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145437117","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}