1
|
Guo C, Xi L, Qu X, Huang Z, Li S, Li W, Liu X, Zhang J. The pharmacokinetic-nephrotoxicity relationships of CMS and CMS-E2 from the perspective of plasma and kidney drug concentrations in rats. Sci Rep 2025; 15:16299. [PMID: 40348891 PMCID: PMC12065821 DOI: 10.1038/s41598-025-96407-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/27/2025] [Indexed: 05/14/2025] Open
Abstract
Nephrotoxicity has seriously affected the clinical application of colistin methanesulphonate (CMS). Colistin B methanesulphonate (CMS-E2) is a novel polymyxin developed and aimed to have lower nephrotoxicity. This study aimed to investigate the relationships between pharmacokinetics (PK) and nephrotoxicity of CMS and CMS-E2 and compare the toxicity of the two drugs in rats. Rats were treated intraperitoneally with a single dose of saline, CMS [10, 20 mg/kg of colistin base activity (CBA)], and CMS-E2 (20, 40 mg/kg CBA). An LC-MS/MS method was developed to determine plasma and renal tissue concentrations of CMS/CMS-E2 and colistin/colistin B. The severity of renal injuries was examined both biochemically and histologically. The PK-toxicodynamic (TD) model was evaluated to characterize the PK of CMS/CMS-E2 and colistin/colistin B in plasma as well as its relationship with nephrotoxicity. Creatinine (CR) and blood urea nitrogen (BUN) profiles were described using an indirect link PK-TD model, with linear-effect relationship. Both the slope between colistin or colistin B concentrations in the effect compartment and CR, BUN was significantly lower for CMS-E2 compared with CMS (CR: P = 0.027, BUN: P = 0.043). The concentrations of colistin and colistin B in kidneys were correlated with CR, BUN values, and histologic examination scores. The regression coefficient of CMS-E2 between the colistin B concentrations in renal tissues and CR, BUN values were lower, as well (CR: P = 0.003, BUN: P = 0.001). The renal injuries induced by CMS and CMS-E2 lagged behind the change of plasma colistin or colistin B concentrations and correlated to those in kidneys. CMS-E2 showed significantly lower nephrotoxicity compared to CMS in vivo.
Collapse
Affiliation(s)
- Chenxue Guo
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai, 200040, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Lin Xi
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai, 200040, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xingyi Qu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai, 200040, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Zhiwei Huang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai, 200040, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Size Li
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai, 200040, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Wanzhen Li
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai, 200040, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xiaofen Liu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, 200040, China.
- Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai, 200040, China.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Jing Zhang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, 200040, China.
- Key Laboratory of Clinical Pharmacology of Antibiotics, Shanghai, 200040, China.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China.
- Clinical Pharmacology Research Center, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
2
|
Mi K, Wu X, Lin Z. Chemical risk assessment in food animals via physiologically based pharmacokinetic modeling - Part I: Veterinary drugs on human food safety assessment. ENVIRONMENT INTERNATIONAL 2025; 197:109339. [PMID: 39986004 DOI: 10.1016/j.envint.2025.109339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/10/2025] [Accepted: 02/16/2025] [Indexed: 02/24/2025]
Abstract
Veterinary drugs and environmental pollutants can enter food animals and remain as residues in food chains threatening human food safety and health. Performing health risk and food safety assessments to derive safety levels of these xenobiotics can protect human health. Physiologically based pharmacokinetic (PBPK) modeling is a mathematical tool to quantitatively describe chemical disposition in humans and animals informing human food safety and health risk assessments. However, few reviews focus on the application of PBPK models in food animals and discuss their relationship to human food safety and health risk assessments in the last five years (2020-2024). In this series of reviews, we introduce the methodology, recent progress and challenges of PBPK modeling in food animals. The present review is Part I of this series of reviews and it focuses on applications of PBPK models of veterinary drugs in food animals, whereas Part II is a companion review focusing on environmental chemicals. Advanced strategies of PBPK modeling in risk and food safety assessment, including population PBPK, interactive PBPK web dashboard, and generic PBPK are also summarized in Part I. Additionally, we share our perspective on the existing challenges and future direction for PBPK modeling of veterinary medicines in food animals.
Collapse
Affiliation(s)
- Kun Mi
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32611, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32611, USA.
| | - Xue Wu
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32611, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32611, USA.
| | - Zhoumeng Lin
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32611, USA; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
3
|
Zhao C, Kristoffersson AN, Khan DD, Lagerbäck P, Lustig U, Cao S, Annerstedt C, Cars O, Andersson DI, Hughes D, Nielsen EI, Friberg LE. Quantifying combined effects of colistin and ciprofloxacin against Escherichia coli in an in silico pharmacokinetic-pharmacodynamic model. Sci Rep 2024; 14:11706. [PMID: 38778123 PMCID: PMC11111785 DOI: 10.1038/s41598-024-61518-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024] Open
Abstract
Co-administering a low dose of colistin (CST) with ciprofloxacin (CIP) may improve the antibacterial effect against resistant Escherichia coli, offering an acceptable benefit-risk balance. This study aimed to quantify the interaction between ciprofloxacin and colistin in an in silico pharmacokinetic-pharmacodynamic model from in vitro static time-kill experiments (using strains with minimum inhibitory concentrations, MICCIP 0.023-1 mg/L and MICCST 0.5-0.75 mg/L). It was also sought to demonstrate an approach of simulating concentrations at the site of infection with population pharmacokinetic and whole-body physiologically based pharmacokinetic models to explore the clinical value of the combination when facing more resistant strains (using extrapolated strains with lower susceptibility). The combined effect in the final model was described as the sum of individual drug effects with a change in drug potency: for ciprofloxacin, concentration at half maximum killing rate (EC50) in combination was 160% of the EC50 in monodrug experiments, while for colistin, the change in EC50 was strain-dependent from 54.1% to 119%. The benefit of co-administrating a lower-than-commonly-administrated colistin dose with ciprofloxacin in terms of drug effect in comparison to either monotherapy was predicted in simulated bloodstream infections and pyelonephritis. The study illustrates the value of pharmacokinetic-pharmacodynamic modelling and simulation in streamlining rational development of antibiotic combinations.
Collapse
Affiliation(s)
- Chenyan Zhao
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | | | - David D Khan
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | | | - Ulrika Lustig
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Sha Cao
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | | | - Otto Cars
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Dan I Andersson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Diarmaid Hughes
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | | | - Lena E Friberg
- Department of Pharmacy, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
4
|
Mi K, Sun L, Hou Y, Cai X, Zhou K, Ma W, Xu X, Pan Y, Liu Z, Huang L. A physiologically based pharmacokinetic model to optimize the dosage regimen and withdrawal time of cefquinome in pigs. PLoS Comput Biol 2023; 19:e1011331. [PMID: 37585381 PMCID: PMC10431683 DOI: 10.1371/journal.pcbi.1011331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 07/06/2023] [Indexed: 08/18/2023] Open
Abstract
Cefquinome is widely used to treat respiratory tract diseases of swine. While extra-label dosages of cefquinome could improve clinical efficacy, they might lead to excessively high residues in animal-derived food. In this study, a physiologically based pharmacokinetic (PBPK) model was calibrated based on the published data and a microdialysis experiment to assess the dosage efficiency and food safety. For the microdialysis experiment, in vitro/in vivo relative recovery and concentration-time curves of cefquinome in the lung interstitium were investigated. This PBPK model is available to predict the drug concentrations in the muscle, kidney, liver, plasma, and lung interstitial fluid. Concentration-time curves of 1000 virtual animals in different tissues were simulated by applying sensitivity and Monte Carlo analyses. By integrating pharmacokinetic/pharmacodynamic target parameters, cefquinome delivered at 3-5 mg/kg twice daily is advised for the effective control of respiratory tract infections of nursery pig, which the bodyweight is around 25 kg. Based on the predicted cefquinome concentrations in edible tissues, the withdrawal interval is 2 and 3 days for label and the extra-label doses, respectively. This study provides a useful tool to optimize the dosage regimen of cefquinome against respiratory tract infections and predicts the concentration of cefquinome residues in edible tissues. This information would be helpful to improve the food safety and guide rational drug usage.
Collapse
Affiliation(s)
- Kun Mi
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and National Safety Laboratory of Veterinary Drug (HZAU), Wuhan, China
- MOA Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, China
| | - Lei Sun
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and National Safety Laboratory of Veterinary Drug (HZAU), Wuhan, China
| | - Yixuan Hou
- MOA Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, China
| | - Xin Cai
- MOA Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, China
| | - Kaixiang Zhou
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Wuhan, China
| | - Wenjin Ma
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiangyue Xu
- MOA Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, China
| | - Yuanhu Pan
- MOA Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, China
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhenli Liu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and National Safety Laboratory of Veterinary Drug (HZAU), Wuhan, China
- MOA Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, China
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Wuhan, China
| | - Lingli Huang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and National Safety Laboratory of Veterinary Drug (HZAU), Wuhan, China
- MOA Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, China
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
5
|
Qi B, Gijsen M, De Vocht T, Deferm N, Van Brantegem P, Abza GB, Nauwelaerts N, Wauters J, Spriet I, Annaert P. Unravelling the Hepatic Elimination Mechanisms of Colistin. Pharm Res 2023; 40:1723-1734. [PMID: 37258948 DOI: 10.1007/s11095-023-03536-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 05/13/2023] [Indexed: 06/02/2023]
Abstract
PURPOSE Colistin is an antibiotic which is increasingly used as a last-resort therapy in critically-ill patients with multidrug resistant Gram-negative infections. The purpose of this study was to evaluate the mechanisms underlying colistin's pharmacokinetic (PK) behavior and to characterize its hepatic metabolism. METHODS In vitro incubations were performed using colistin sulfate with rat liver microsomes (RLM) and with rat and human hepatocytes (RH and HH) in suspension. The uptake of colistin in RH/HH and thefraction of unbound colistin in HH (fu,hep) was determined. In vitro to in vivo extrapolation (IVIVE) was employed to predict the hepatic clearance (CLh) of colistin. RESULTS Slow metabolism was detected in RH/HH, with intrinsic clearance (CLint) values of 9.34± 0.50 and 3.25 ± 0.27 mL/min/kg, respectively. Assuming the well-stirred model for hepatic drug elimination, the predicted rat CLh was 3.64± 0.22 mL/min/kg which could explain almost 70% of the reported non-renal in vivo clearance. The predicted human CLh was 91.5 ± 8.83 mL/min, which was within two-fold of the reported plasma clearance in healthy volunteers. When colistin was incubated together with the multidrug resistance-associated protein (MRP/Mrp) inhibitor benzbromarone, the intracellular accumulation of colistin in RH/HH increased significantly. CONCLUSION These findings indicate the major role of hepatic metabolism in the non-renal clearance of colistin, while MRP/Mrp-mediated efflux is involved in the hepatic disposition of colistin. Our data provide detailed quantitative insights into the hereto unknown mechanisms responsible for non-renal elimination of colistin.
Collapse
Affiliation(s)
- Bing Qi
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
- The Second Affiliated Hospital, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Matthias Gijsen
- Clinical Pharmacology and Pharmacotherapy, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
- Pharmacy Department, University Hospitals Leuven, Leuven, Belgium
| | - Tom De Vocht
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Neel Deferm
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Pieter Van Brantegem
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Getahun B Abza
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Nina Nauwelaerts
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Joost Wauters
- Clinical Infectious and Inflammatory Disorders, KU Leuven Department of Microbiology and Immunology; Medical Intensive Care Unit, University Hospitals Leuven, Leuven, Belgium
| | - Isabel Spriet
- Clinical Pharmacology and Pharmacotherapy, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
- Pharmacy Department, University Hospitals Leuven, Leuven, Belgium
| | - Pieter Annaert
- Drug Delivery and Disposition, KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium.
| |
Collapse
|
6
|
Zhu S, Zhang J, Lv Z, Zhu P, Oo C, Yu M, Sy SKB. Prediction of Tissue Exposures of Meropenem, Colistin, and Sulbactam in Pediatrics Using Physiologically Based Pharmacokinetic Modeling. Clin Pharmacokinet 2022; 61:1427-1441. [PMID: 35947360 DOI: 10.1007/s40262-022-01161-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2022] [Indexed: 01/10/2023]
Abstract
BACKGROUND The combination of polymyxins, meropenem, and sulbactam demonstrated efficacy against multi-drug-resistant bacillus Acinetobacter baumannii. These three antibiotics are commonly used against major blood, skin, lung, and heart muscle infections. OBJECTIVE The objective of this study was to predict drug disposition and extrapolate the efficacy in these tissues using a physiologically based pharmacokinetic modeling approach that linked drug exposures to their target pharmacodynamic indices associated with antimicrobial activities against A. baumannii. METHODS An adult physiologically based pharmacokinetic model was developed for meropenem, colistin, and sulbactam and scaled to pediatrics accounting for both renal and non-renal clearances. The model reliability was evaluated by comparing simulated plasma and tissue drug exposures to observed data. Target pharmacodynamic indices were used to evaluate whether pediatric and adult dosing regimens provided sufficient coverage. RESULTS The modeled plasma drug exposures in adults and pediatric patients were consistent with reported literature data. The mean fold errors for meropenem, colistin, and sulbactam were in the range of 0.710-1.37, 0.981-1.47, and 0.647-1.39, respectively. Simulated exposures in the blood, skin, lung, and heart were consistent with reported penetration rates. In a virtual pediatric population aged from 2 to < 18 years, the interpretive breakpoints were achieved in 85-90% of subjects for their targeted pharmacodynamic indices after administration of pediatric dosing regimens consisting of 30 mg/kg of meropenem, and 40 mg/kg of sulbactam three times daily as a 3-h or continuous infusion and 5 mg/kg/day of colistin base activity. CONCLUSIONS The physiologically based pharmacokinetic modeling supports pediatric dosing regimens of meropenem/colistin/sulbactam in a co-administration setting against infections in the blood, lung, skin, and heart tissues due to A. baumannii.
Collapse
Affiliation(s)
- Shixing Zhu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, People's Republic of China
| | - Jiayuan Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, People's Republic of China
| | - Zhihua Lv
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, People's Republic of China.,Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, People's Republic of China
| | - Peijuan Zhu
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, USA
| | - Charles Oo
- SunLife Biopharma, Morris Plains, NJ, USA
| | - Mingming Yu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, People's Republic of China. .,Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, People's Republic of China.
| | - Sherwin K B Sy
- Department of Statistics, State University of Maringá, Maringá, Paraná, Brazil.
| |
Collapse
|
7
|
Lipp MA, Crass RL, Fitzgerald LJ, Patel TS, Simon RH, Lenhan BE, Han MK, Jia S. Acute kidney injury in cystic fibrosis patients treated with intravenous colistimethate sodium or tobramycin. J Antimicrob Chemother 2022; 77:2516-2521. [PMID: 35678460 DOI: 10.1093/jac/dkac187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 05/17/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Colistimethate sodium and tobramycin are important systemic antibiotics for treatment of cystic fibrosis (CF) pulmonary exacerbations but can induce acute kidney injury (AKI). We characterize the rate of AKI in CF patients treated with systemic colistimethate sodium compared with tobramycin. METHODS This single-centre, retrospective cohort study included hospitalized CF patients treated with IV colistimethate sodium or tobramycin. The primary outcome was AKI defined using the RIFLE criteria. Multivariate logistic regression using a mixed model was performed to identify variables that were independently associated with AKI. RESULTS Overall, 156 patients representing 507 care encounters were included. The OR of AKI was not increased with IV colistimethate sodium relative to IV tobramycin after adjusting for other potential predictor variables (aOR 1.00; 95% CI 0.16-6.03). The frequency of AKI was 9.5% across all encounters, 6.9% with IV colistimethate sodium and 9.9% with IV tobramycin, with RIFLE category R (risk) being the most common stage, accounting for 4.2% of encounters with IV colistimethate sodium and 9.2% with IV tobramycin. The concomitant use of another nephrotoxin (aOR 2.51; 95% CI 1.27-4.95) or the combination of vancomycin and piperacillin/tazobactam (aOR 5.95; 95% CI 2.05-17.3) were both associated with increased odds of AKI. CONCLUSIONS Systemic treatment with colistimethate sodium or tobramycin in the CF patient population is associated with a similar rate of nephrotoxicity. However, clinicians should be mindful of the increased risk for AKI in patients treated with either IV colistimethate sodium or IV tobramycin when used concurrently with other nephrotoxic agents, particularly the combination of vancomycin and piperacillin/tazobactam.
Collapse
Affiliation(s)
- Madeline A Lipp
- Department of Clinical Pharmacy and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ryan L Crass
- Ann Arbor Pharmacometrics Group, Ann Arbor, MI, USA
| | | | - Twisha S Patel
- Department of Pharmacy Services, Michigan Medicine, Ann Arbor, MI, USA
| | - Richard H Simon
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Blair E Lenhan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - MeiLan K Han
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Shijing Jia
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
8
|
Jarzina S, Di Fiore S, Ellinger B, Reiser P, Frank S, Glaser M, Wu J, Taverne FJ, Kramer NI, Mally A. Application of the Adverse Outcome Pathway Concept to In Vitro Nephrotoxicity Assessment: Kidney Injury due to Receptor-Mediated Endocytosis and Lysosomal Overload as a Case Study. FRONTIERS IN TOXICOLOGY 2022; 4:864441. [PMID: 35516525 PMCID: PMC9061999 DOI: 10.3389/ftox.2022.864441] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/22/2022] [Indexed: 11/20/2022] Open
Abstract
Application of adverse outcome pathways (AOP) and integration of quantitative in vitro to in vivo extrapolation (QIVIVE) may support the paradigm shift in toxicity testing to move from apical endpoints in test animals to more mechanism-based in vitro assays. Here, we developed an AOP of proximal tubule injury linking a molecular initiating event (MIE) to a cascade of key events (KEs) leading to lysosomal overload and ultimately to cell death. This AOP was used as a case study to adopt the AOP concept for systemic toxicity testing and risk assessment based on in vitro data. In this AOP, nephrotoxicity is thought to result from receptor-mediated endocytosis (MIE) of the chemical stressor, disturbance of lysosomal function (KE1), and lysosomal disruption (KE2) associated with release of reactive oxygen species and cytotoxic lysosomal enzymes that induce cell death (KE3). Based on this mechanistic framework, in vitro readouts reflecting each KE were identified. Utilizing polymyxin antibiotics as chemical stressors for this AOP, the dose-response for each in vitro endpoint was recorded in proximal tubule cells from rat (NRK-52E) and human (RPTEC/TERT1) in order to (1) experimentally support the sequence of key events (KEs), to (2) establish quantitative relationships between KEs as a basis for prediction of downstream KEs based on in vitro data reflecting early KEs and to (3) derive suitable in vitro points of departure for human risk assessment. Time-resolved analysis was used to support the temporal sequence of events within this AOP. Quantitative response-response relationships between KEs established from in vitro data on polymyxin B were successfully used to predict in vitro toxicity of other polymyxin derivatives. Finally, a physiologically based kinetic (PBK) model was utilized to transform in vitro effect concentrations to a human equivalent dose for polymyxin B. The predicted in vivo effective doses were in the range of therapeutic doses known to be associated with a risk for nephrotoxicity. Taken together, these data provide proof-of-concept for the feasibility of in vitro based risk assessment through integration of mechanistic endpoints and reverse toxicokinetic modelling.
Collapse
Affiliation(s)
| | - Stefano Di Fiore
- Fraunhofer Institute for Molecular Biology and Applied Ecology, Division Molecular Biotechnology Aachen, Aachen, Germany
| | - Bernhard Ellinger
- Fraunhofer Institute for Molecular Biology and Applied Ecology, Division Translational Medicine, ScreeningPort, Hamburg, Germany
| | - Pia Reiser
- Department of Toxicology, University of Würzburg, Würzburg, Germany
| | - Sabrina Frank
- Department of Toxicology, University of Würzburg, Würzburg, Germany
| | - Markus Glaser
- Department of Toxicology, University of Würzburg, Würzburg, Germany
| | - Jiaqing Wu
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, Netherlands
- Toxicology Division, Wageningen University, Wageningen, Netherlands
| | - Femke J. Taverne
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, Netherlands
- Host-microbe Interactions, Wageningen University, Wageningen, Netherlands
| | - Nynke I. Kramer
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, Netherlands
- Toxicology Division, Wageningen University, Wageningen, Netherlands
| | - Angela Mally
- Department of Toxicology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
9
|
Nebulized Colistin in Ventilator-Associated Pneumonia and Tracheobronchitis: Historical Background, Pharmacokinetics and Perspectives. Microorganisms 2021; 9:microorganisms9061154. [PMID: 34072189 PMCID: PMC8227626 DOI: 10.3390/microorganisms9061154] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 05/14/2021] [Accepted: 05/20/2021] [Indexed: 11/17/2022] Open
Abstract
Clinical evidence suggests that nebulized colistimethate sodium (CMS) has benefits for treating lower respiratory tract infections caused by multidrug-resistant Gram-negative bacteria (GNB). Colistin is positively charged, while CMS is negatively charged, and both have a high molecular mass and are hydrophilic. These physico-chemical characteristics impair crossing of the alveolo-capillary membrane but enable the disruption of the bacterial wall of GNB and the aggregation of the circulating lipopolysaccharide. Intravenous CMS is rapidly cleared by glomerular filtration and tubular excretion, and 20-25% is spontaneously hydrolyzed to colistin. Urine colistin is substantially reabsorbed by tubular cells and eliminated by biliary excretion. Colistin is a concentration-dependent antibiotic with post-antibiotic and inoculum effects. As CMS conversion to colistin is slower than its renal clearance, intravenous administration can lead to low plasma and lung colistin concentrations that risk treatment failure. Following nebulization of high doses, colistin (200,000 international units/24h) lung tissue concentrations are > five times minimum inhibitory concentration (MIC) of GNB in regions with multiple foci of bronchopneumonia and in the range of MIC breakpoints in regions with confluent pneumonia. Future research should include: (1) experimental studies using lung microdialysis to assess the PK/PD in the interstitial fluid of the lung following nebulization of high doses of colistin; (2) superiority multicenter randomized controlled trials comparing nebulized and intravenous CMS in patients with pandrug-resistant GNB ventilator-associated pneumonia and ventilator-associated tracheobronchitis; (3) non-inferiority multicenter randomized controlled trials comparing nebulized CMS to intravenous new cephalosporines/ß-lactamase inhibitors in patients with extensive drug-resistant GNB ventilator-associated pneumonia and ventilator-associated tracheobronchitis.
Collapse
|
10
|
Kristoffersson AN, Rognås V, Brill MJE, Dishon-Benattar Y, Durante-Mangoni E, Daitch V, Skiada A, Lellouche J, Nutman A, Kotsaki A, Andini R, Eliakim-Raz N, Bitterman R, Antoniadou A, Karlsson MO, Theuretzbacher U, Leibovici L, Daikos GL, Mouton JW, Carmeli Y, Paul M, Friberg LE. Population pharmacokinetics of colistin and the relation to survival in critically ill patients infected with colistin susceptible and carbapenem-resistant bacteria. Clin Microbiol Infect 2020; 26:1644-1650. [PMID: 32213316 DOI: 10.1016/j.cmi.2020.03.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 02/26/2020] [Accepted: 03/15/2020] [Indexed: 02/08/2023]
Abstract
OBJECTIVES The aim was to analyse the population pharmacokinetics of colistin and to explore the relationship between colistin exposure and time to death. METHODS Patients included in the AIDA randomized controlled trial were treated with colistin for severe infections caused by carbapenem-resistant Gram-negative bacteria. All subjects received a 9 million units (MU) loading dose, followed by a 4.5 MU twice daily maintenance dose, with dose reduction if creatinine clearance (CrCL) < 50 mL/min. Individual colistin exposures were estimated from the developed population pharmacokinetic model and an optimized two-sample per patient sampling design. Time to death was evaluated in a parametric survival analysis. RESULTS Out of 406 randomized patients, 349 contributed pharmacokinetic data. The median (90% range) colistin plasma concentration was 0.44 (0.14-1.59) mg/L at 15 minutes after the end of first infusion. In samples drawn 10 hr after a maintenance dose, concentrations were >2 mg/L in 94% (195/208) and 44% (38/87) of patients with CrCL ≤120 mL/min, and >120 mL/min, respectively. Colistin methanesulfonate sodium (CMS) and colistin clearances were strongly dependent on CrCL. High colistin exposure to MIC ratio was associated with increased hazard of death in the multivariate analysis (adjusted hazard ratio (95% CI): 1.07 (1.03-1.12)). Other significant predictors included SOFA score at baseline (HR 1.24 (1.19-1.30) per score increase), age and Acinetobacter or Pseudomonas as index pathogen. DISCUSSION The population pharmacokinetic model predicted that >90% of the patients had colistin concentrations >2 mg/L at steady state, but only 66% at 4 hr after start of treatment. High colistin exposure was associated with poor kidney function, and was not related to a prolonged survival.
Collapse
Affiliation(s)
- A N Kristoffersson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - V Rognås
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - M J E Brill
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Y Dishon-Benattar
- Institute of Infectious Diseases, Rambam Health Care Campus, Haifa, Israel; The Cheryl Spencer Institute for Nursing Research, University of Haifa, Israel
| | - E Durante-Mangoni
- Department of Precision Medicine, University of Campania 'L Vanvitelli' and AORN dei Colli-Monaldi Hospital, Napoli, Italy
| | - V Daitch
- Infectious Diseases University Research Centre, Rabin Medical Centre, Beilinson Hospital, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel-Aviv University, and Department of Medicine E, Rabin Medical Centre, Beilinson Hospital, Petah Tikva, Israel
| | - A Skiada
- First Department of Medicine, Laiko General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - J Lellouche
- National Centre for Infection Control and Antibiotic Resistance, Tel Aviv Medical Centre, Tel Aviv, Israel; National Laboratory for Antibiotic Resistance and Investigation of Outbreaks in Medical Institutions, Tel Aviv Medical Centre, Tel Aviv, Israel
| | - A Nutman
- National Centre for Infection Control and Antibiotic Resistance, Tel Aviv Medical Centre, Tel Aviv, Israel
| | - A Kotsaki
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, School of Medicine, University General Hospital Attikon, Athens, Greece
| | - R Andini
- Department of Precision Medicine, University of Campania 'L Vanvitelli' and AORN dei Colli-Monaldi Hospital, Napoli, Italy
| | - N Eliakim-Raz
- Infectious Diseases University Research Centre, Rabin Medical Centre, Beilinson Hospital, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel-Aviv University, and Department of Medicine E, Rabin Medical Centre, Beilinson Hospital, Petah Tikva, Israel
| | - R Bitterman
- Institute of Infectious Diseases, Rambam Health Care Campus, Haifa, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Techion - Israel Institute of Technology, Haifa, Israel
| | - A Antoniadou
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, School of Medicine, University General Hospital Attikon, Athens, Greece
| | - M O Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | | - L Leibovici
- Sackler Faculty of Medicine, Tel-Aviv University, and Department of Medicine E, Rabin Medical Centre, Beilinson Hospital, Petah Tikva, Israel; Department of Medicine E, Rabin Medical Centre, Beilinson Hospital, Petah Tikva, Israel
| | - G L Daikos
- First Department of Medicine, Laiko General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - J W Mouton
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, Rotterdam, the Netherlands
| | - Y Carmeli
- National Centre for Infection Control and Antibiotic Resistance, Tel Aviv Medical Centre, Tel Aviv, Israel; National Laboratory for Antibiotic Resistance and Investigation of Outbreaks in Medical Institutions, Tel Aviv Medical Centre, Tel Aviv, Israel
| | - M Paul
- Institute of Infectious Diseases, Rambam Health Care Campus, Haifa, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Techion - Israel Institute of Technology, Haifa, Israel
| | - L E Friberg
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
11
|
Pharmacokinetics of Polymyxins in Animals. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1145:89-103. [DOI: 10.1007/978-3-030-16373-0_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
12
|
Bouchene S, Marchand S, Couet W, Friberg LE, Gobin P, Lamarche I, Grégoire N, Björkman S, Karlsson MO. A Whole-Body Physiologically Based Pharmacokinetic Model for Colistin and Colistin Methanesulfonate in Rat. Basic Clin Pharmacol Toxicol 2018; 123:407-422. [PMID: 29665289 DOI: 10.1111/bcpt.13026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/03/2018] [Indexed: 11/29/2022]
Abstract
Colistin is a polymyxin antibiotic used to treat patients infected with multidrug-resistant Gram-negative bacteria (MDR-GNB). The objective of this work was to develop a whole-body physiologically based pharmacokinetic (WB-PBPK) model to predict tissue distribution of colistin in rat. The distribution of a drug in a tissue is commonly characterized by its tissue-to-plasma partition coefficient, Kp . Colistin and its prodrug, colistin methanesulfonate (CMS) Kp priors, were measured experimentally from rat tissue homogenates or predicted in silico. The PK parameters of both compounds were estimated fitting in vivo their plasma concentration-time profiles from six rats receiving an i.v. bolus of CMS. The variability in the data was quantified by applying a nonlinear mixed effect (NLME) modelling approach. A WB-PBPK model was developed assuming a well-stirred and perfusion-limited distribution in tissue compartments. Prior information on tissue distribution of colistin and CMS was investigated following three scenarios: Kp was estimated using in silico Kp priors (I) or Kp was estimated using experimental Kp priors (II) or Kp was fixed to the experimental values (III). The WB-PBPK model best described colistin and CMS plasma concentration-time profiles in scenario II. Colistin-predicted concentrations in kidneys in scenario II were higher than in other tissues, which was consistent with its large experimental Kp prior. This might be explained by a high affinity of colistin for renal parenchyma and active reabsorption into the proximal tubular cells. In contrast, renal accumulation of colistin was not predicted in scenario I. Colistin and CMS clearance estimates were in agreement with published values. The developed model suggests using experimental priors over in silico Kp priors for kidneys to provide a better prediction of colistin renal distribution. Such models might serve in drug development for interspecies scaling and investigate the impact of disease state on colistin disposition.
Collapse
Affiliation(s)
- Salim Bouchene
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Sandrine Marchand
- INSERM U-1070, Pôle Biologie Santé, Poitiers, France.,Laboratoire de Toxicologie et Pharmacocinétique, CHU de Poitiers, Poitiers, France
| | - William Couet
- INSERM U-1070, Pôle Biologie Santé, Poitiers, France.,Laboratoire de Toxicologie et Pharmacocinétique, CHU de Poitiers, Poitiers, France
| | - Lena E Friberg
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Patrice Gobin
- INSERM U-1070, Pôle Biologie Santé, Poitiers, France
| | | | - Nicolas Grégoire
- INSERM U-1070, Pôle Biologie Santé, Poitiers, France.,Laboratoire de Toxicologie et Pharmacocinétique, CHU de Poitiers, Poitiers, France
| | - Sven Björkman
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Mats O Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|