1
|
Hickok NJ, Li B, Oral E, Zaat SAJ, Armbruster DA, Atkins GJ, Chen AF, Coraça-Huber DC, Dai T, Greenfield EM, Kasinath R, Libera M, Marques CNH, Moriarty TF, Scott Phillips K, Raghuraman K, Ren D, Rimondini L, Saeed K, Schaer TP, Schwarz EM, Spiegel C, Stoodley P, Truong VK, Tsang STJ, Wildemann B, Zelmer AR, Zinkernagel AS. The 2023 Orthopedic Research Society's international consensus meeting on musculoskeletal infection: Summary from the in vitro section. J Orthop Res 2024; 42:512-517. [PMID: 38146070 DOI: 10.1002/jor.25774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/21/2023] [Indexed: 12/27/2023]
Abstract
Antimicrobial strategies for musculoskeletal infections are typically first developed with in vitro models. The In Vitro Section of the 2023 Orthopedic Research Society Musculoskeletal Infection international consensus meeting (ICM) probed our state of knowledge of in vitro systems with respect to bacteria and biofilm phenotype, standards, in vitro activity, and the ability to predict in vivo efficacy. A subset of ICM delegates performed systematic reviews on 15 questions and made recommendations and assessment of the level of evidence that were then voted on by 72 ICM delegates. Here, we report recommendations and rationale from the reviews and the results of the internet vote. Only two questions received a ≥90% consensus vote, emphasizing the disparate approaches and lack of established consensus for in vitro modeling and interpretation of results. Comments on knowledge gaps and the need for further research on these critical MSKI questions are included.
Collapse
Affiliation(s)
- Noreen J Hickok
- Department of Orthopaedic Surgery, Department of Biochemistry & Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Bingyun Li
- Department of Orthopaedics, School of Medicine, West Virginia University, Morgantown, West Virginia, USA
| | - Ebru Oral
- Harris Orthopaedic Laboratory, Department of Orthopaedic Surgery, Harvard Medical School, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sebastian A J Zaat
- Department of Medical Microbiology and Infection Prevention, Amsterdam Institute for Infection and Immunity, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Gerald J Atkins
- Centre for Orthopaedic and Trauma Research, University of Adelaide, Adelaide, South Australia, Australia
| | - Antonia F Chen
- Department of Orthopaedics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Débora C Coraça-Huber
- Research Laboratory for Implant Associated Infections (Biofilm Lab), University Hospital for Orthopedics and Traumatology, Experimental Orthopaedics, Medical University of Innsbruck, Innsbruck, Austria
| | - Tianhong Dai
- Harvard Medical School, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Edward M Greenfield
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indiana Center for Musculoskeletal Health, Indianapolis, Indiana, USA
| | | | - Matthew Libera
- Department of Chemical Engineering and Materials Science, Stevens Institute of Technology, Hoboken, New Jersey, USA
| | - Cláudia N H Marques
- Department of Biological Sciences, Binghamton Biofilm Research Center, Binghamton University, Binghamton, New York, USA
| | | | - K Scott Phillips
- Laboratory of Analytical Chemistry, Division of Biological Standards and Quality Control, Office of Compliance and Biologics Quality, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | | | - Dacheng Ren
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York, USA
| | - Lia Rimondini
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Kordo Saeed
- University Hospital Southampton NHS Foundation Trust, Winchester and Basingstoke, UK
- University of Southampton, Southampton, UK
| | - Thomas P Schaer
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania, Kennett Square, Pennsylvania, USA
| | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester, Rochester, New York, USA
| | - Christopher Spiegel
- Research Laboratory for Biofilms and Implant Associated Infections (BIOFILM LAB), Experimental Orthopedics, University Hospital for Orthopedics and Traumatology, Medical University of Innsbruck, Innsbruck, Austria
| | - Paul Stoodley
- Department Microbial Infection and Immunity and Department of Orthopaedics, The Ohio State University, Columbus, Ohio, USA
| | - Vi Khanh Truong
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Bedford Park, South Australia, Australia
| | - Shao-Ting Jerry Tsang
- Department of Trauma and Orthopaedic Surgery, University of Edinburgh, Edinburgh, Scotland, UK
| | - Britt Wildemann
- Experimental Trauma Surgery, Department of Trauma, Hand and Reconstructive Surgery, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Anja R Zelmer
- Centre for Orthopaedic and Trauma Research, University of Adelaide, Adelaide, South Australia, Australia
| | - Annelies S Zinkernagel
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital of Zurich and University of Zurich, Zurich, Switzerland
| |
Collapse
|
2
|
Minichmayr IK, Aranzana-Climent V, Friberg LE. Pharmacokinetic-pharmacodynamic models for time courses of antibiotic effects: VSI: Antimicrobial Pharmacometrics. Int J Antimicrob Agents 2022; 60:106616. [PMID: 35691605 DOI: 10.1016/j.ijantimicag.2022.106616] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 05/18/2022] [Accepted: 05/29/2022] [Indexed: 11/16/2022]
Abstract
Pharmacokinetic-pharmacodynamic (PKPD) models have emerged as valuable tools for the characterisation and translation of antibiotic effects, and consequently for drug development and therapy. In contrast to traditional PKPD concepts for antibiotics like MIC and PKPD indices, PKPD models enable to describe the continuous, often species- or population-dependent time course of antimicrobial effects, commonly considering mechanistic pathogen- and drug-related knowledge. This review presents a comprehensive overview of previously published PKPD models describing repeated measurements of antibiotic effects. We conducted a literature review to identify PKPD models based on (i) antibiotic compounds, (ii) Gram-positive or Gram-negative pathogens, and (iii) in vitro or in vivo longitudinal colony forming unit data. We identified 132 publications released between 1963 and 2021, including models based on exposure with single antibiotics (n=92) and drug combinations (n=40), as well as different experimental settings (e.g., static/traditional dynamic/hollow-fibre/animal time-kill models, n=90/27/32/11). An interactive, fully searchable table summarises the details of each model, i.e. variants and mechanistic elements of PKPD submodels capturing observed bacterial growth, regrowth, drug effects, and interactions. Furthermore, the review highlights main purposes of PKPD model development, including the translation of preclinical PKPD to clinical settings and the assessment of varied dosing regimens and patient characteristics for their impact on clinical antibiotic effects. In summary, this comprehensive overview of PKPD models shall assist in identifying PKPD modelling strategies to describe growth, killing, regrowth and interaction patterns for pathogen-antibiotic combinations over time and ultimately facilitate model-informed antibiotic translation, dosing and drug development.
Collapse
Affiliation(s)
- Iris K Minichmayr
- Department of Pharmacy, Uppsala University, Box 580, 75123 Uppsala, Sweden
| | | | - Lena E Friberg
- Department of Pharmacy, Uppsala University, Box 580, 75123 Uppsala, Sweden.
| |
Collapse
|
3
|
Optimization and Validation of Dosage Regimen for Ceftiofur against Pasteurella multocida in Swine by Physiological Based Pharmacokinetic-Pharmacodynamic Model. Int J Mol Sci 2022; 23:ijms23073722. [PMID: 35409082 PMCID: PMC8998519 DOI: 10.3390/ijms23073722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/10/2022] [Accepted: 03/12/2022] [Indexed: 12/10/2022] Open
Abstract
Model informed drug development is a valuable tool for drug development and clinical application due to its ability to integrate variability and uncertainty of data. This study aimed to determine an optimal dosage of ceftiofur against P. multocida by ex vivo pharmacokinetic/pharmacodynamic (PK/PD) model and validate the dosage regimens by Physiological based Pharmacokinetic-Pharmacodynamic (PBPK/PD) model. The pharmacokinetic profiles of ceftiofur both in plasma and bronchoalveolar lavage fluid (BALF) are determined. PD performance of ceftiofur against P. multocida was investigated. By establishing PK/PD model, PK/PD parameters and doses were determined. PBPK model and PBPK/PD model were developed to validate the dosage efficacy. The PK/PD parameters, AUC0–24 h/MIC, for bacteriostatic action, bactericidal action and elimination were determined as 44.02, 89.40, and 119.90 h and the corresponding dosages were determined as 0.22, 0.46, and 0.64 mg/kg, respectively. AUC24 h/MIC and AUC 72 h/MIC are simulated by PBPK model, compared with the PK/PD parameters, the therapeutic effect can reach probability of target attainment (PTA) of 90%. The time-courses of bacterial growth were predicted by the PBPK/PD model, which indicated the dosage of 0.46 mg/kg body weight could inhibit the bacterial growth and perform good bactericidal effect.
Collapse
|
4
|
Uropathogenic Escherichia coli Shows Antibiotic Tolerance and Growth Heterogeneity in an In Vitro Model of Intracellular Infection. Antimicrob Agents Chemother 2021; 65:e0146821. [PMID: 34570646 DOI: 10.1128/aac.01468-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Uropathogenic Escherichia coli (UPEC), the major causative agent of urinary tract infections, can invade different types of host cells. To compare the pharmacodynamic properties of antibiotics against intra- and extracellular UPEC, an in vitro model of intracellular infection was established in J774 mouse macrophages infected by the UPEC strain CFT073. We tested antibiotics commonly prescribed against urinary tract infections (gentamicin, ampicillin, nitrofurantoin, trimethoprim, sulfamethoxazole, and ciprofloxacin) and the investigational fluoroquinolone finafloxacin. The metabolic activity of individual bacteria was assessed by expressing the fluorescent reporter protein TIMERbac within CFT073. Concentration-response experiments revealed that all tested antibiotics were much less effective against intracellular bacteria than extracellular ones. Most antibiotics, except fluoroquinolones, were unable to reach a bactericidal effect intracellularly at clinically achievable concentrations. Ciprofloxacin and finafloxacin killed 99.9% of extracellular bacteria at concentrations around the MIC, while for intracellular bacteria, concentrations more than 100× over the MIC were required to achieve a bactericidal effect. Time-kill curves showed that finafloxacin was more rapidly bactericidal in acidic medium than at neutral pH, while the reverse observation was made for ciprofloxacin. Intracellularly, kill curves showed biphasic kinetics for both fluoroquinolones, suggesting the presence of drug-tolerant subpopulations. Flow cytometry analysis of TIMERbac fluorescence revealed a marked heterogeneity in intracellular growth of individual bacteria, suggesting that the presence of subpopulations reaching a state of metabolic dormancy was the main reason for increased antibiotic tolerance of intracellular UPEC.
Collapse
|
5
|
Landersdorfer CB, Nation RL. Limitations of Antibiotic MIC-Based PK-PD Metrics: Looking Back to Move Forward. Front Pharmacol 2021; 12:770518. [PMID: 34776982 PMCID: PMC8585766 DOI: 10.3389/fphar.2021.770518] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 10/14/2021] [Indexed: 12/28/2022] Open
Abstract
Within a few years after the first successful clinical use of penicillin, investigations were conducted in animal infection models to explore a range of factors that were considered likely to influence the antibacterial response to the drug. Those studies identified that the response was influenced by not only the total daily dose but also the interval between individual doses across the day, and whether penicillin was administered in an intermittent or continuous manner. Later, as more antibiotics were discovered and developed, antimicrobial pharmacologists began to measure antibiotic concentrations in biological fluids. This enabled the linking of antibacterial response at a single time point in an animal or in vitro infection model with one of three summary pharmacokinetic (PK) measures of in vivo exposure to the antibiotic. The summary PK exposure measures were normalised to the minimum inhibitory concentration (MIC), an in vitro measure of the pharmacodynamic (PD) potency of the drug. The three PK-PD indices (ratio of maximum concentration to MIC, ratio of area under the concentration-time curve to MIC, time concentration is above MIC) have been used extensively since the 1980s. While these MIC-based summary PK-PD metrics have undoubtedly facilitated the development of new antibiotics and the clinical application of both new and old antibiotics, it is increasingly recognised that they have a number of substantial limitations. In this article we use a historical perspective to review the origins of the three traditional PK-PD indices before exploring in detail their limitations and the implications arising from those limitations. Finally, in the interests of improving antibiotic development and dosing in patients, we consider a model-based approach of linking the full time-course of antibiotic concentrations with that of the antibacterial response. Such an approach enables incorporation of other factors that can influence treatment outcome in patients and has the potential to drive model-informed precision dosing of antibiotics into the future.
Collapse
Affiliation(s)
- Cornelia B Landersdorfer
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Roger L Nation
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| |
Collapse
|
6
|
Seeger J, Guenther S, Schaufler K, Heiden SE, Michelet R, Kloft C. Novel Pharmacokinetic/Pharmacodynamic Parameters Quantify the Exposure-Effect Relationship of Levofloxacin against Fluoroquinolone-Resistant Escherichia coli. Antibiotics (Basel) 2021; 10:antibiotics10060615. [PMID: 34063980 PMCID: PMC8224043 DOI: 10.3390/antibiotics10060615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 11/30/2022] Open
Abstract
Minimal inhibitory concentration-based pharmacokinetic/pharmacodynamic (PK/PD) indices are commonly applied to antibiotic dosing optimisation, but their informative value is limited, as they do not account for bacterial growth dynamics over time. We aimed to comprehensively characterise the exposure–effect relationship of levofloxacin against Escherichia coli and quantify strain-specific characteristics applying novel PK/PD parameters. In vitro infection model experiments were leveraged to explore the exposure–effect relationship of three clinical Escherichia coli isolates, harbouring different genomic fluoroquinolone resistance mechanisms, under constant levofloxacin concentrations or human concentration–time profiles (≤76 h). As an exposure metric, the ‘cumulative area under the levofloxacin–concentration time curve’ was determined. The antibiotic effect was assessed as the ‘cumulative area between the growth control and the bacterial-killing and -regrowth curve’. PK/PD modelling was applied to characterise the exposure–effect relationship and derive novel PK/PD parameters. A sigmoidal Emax model with an inhibition term best characterised the exposure–effect relationship and allowed for discrimination between two isolates sharing the same MIC value. Strain- and exposure-pattern-dependent differences were captured by the PK/PD parameters and elucidated the contribution of phenotypic adaptation to bacterial regrowth. The novel exposure and effect metrics and derived PK/PD parameters allowed for comprehensive characterisation of the isolates and could be applied to overcome the limitations of the MIC in clinical antibiotic dosing decisions, drug research and preclinical development.
Collapse
Affiliation(s)
- Johanna Seeger
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Kelchstr. 31, 12169 Berlin, Germany; (J.S.); (R.M.)
| | - Sebastian Guenther
- Department of Pharmaceutical Biology, Institute of Pharmacy, Universitaet Greifswald, Friedrich-Ludwig-Jahn-Straße 17, 17489 Greifswald, Germany;
| | - Katharina Schaufler
- Department of Pharmaceutical Microbiology, Institute of Pharmacy, Universitaet Greifswald, Friedrich-Ludwig-Jahn-Straße 17, 17489 Greifswald, Germany; (K.S.); (S.E.H.)
| | - Stefan E. Heiden
- Department of Pharmaceutical Microbiology, Institute of Pharmacy, Universitaet Greifswald, Friedrich-Ludwig-Jahn-Straße 17, 17489 Greifswald, Germany; (K.S.); (S.E.H.)
| | - Robin Michelet
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Kelchstr. 31, 12169 Berlin, Germany; (J.S.); (R.M.)
| | - Charlotte Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Kelchstr. 31, 12169 Berlin, Germany; (J.S.); (R.M.)
- Correspondence: ; Tel.: +49-30-838-50656
| |
Collapse
|