Logo: to the web site of Uppsala University

uu.sePublications from Uppsala University
Change search
Link to record
Permanent link

Direct link
Alternative names
Publications (10 of 197) Show all publications
O'Jeanson, A., Nielsen, E. I. & Friberg, L. E. (2025). A model-based evaluation of the pharmacokinetics-pharmacodynamics (PKPD) of avibactam in combination with ceftazidime. JAC - Antimicrobial Resistance, 7(2), Article ID dlaf036.
Open this publication in new window or tab >>A model-based evaluation of the pharmacokinetics-pharmacodynamics (PKPD) of avibactam in combination with ceftazidime
2025 (English)In: JAC - Antimicrobial Resistance, E-ISSN 2632-1823, Vol. 7, no 2, article id dlaf036Article in journal (Refereed) Published
Abstract [en]

Background

The emergence of β-lactamase-producing bacteria limits the effectiveness of β-lactam (BL) antibiotics, and the combination with a β-lactamase inhibitor (BLI) aims to counteract this resistance. However, existing guidelines primarily focus on optimizing the dosing of BLs and do not adequately address the interaction between BLs and BLIs, leading to uncertain pharmacokinetic/pharmacodynamic (PK/PD) targets and potentially suboptimal dosing strategies.

Objectives

To investigate optimal PK/PD targets and dosing strategies for avibactam (BLI) combined with ceftazidime (BL) using mechanism-based PKPD models.

Methods

PK models for ceftazidime and avibactam were integrated with mechanism-based PKPD models for Gram-negative bacteria. Simulations explored dose regimens in mice and humans, evaluating PK/PD indices and computing the PTA for diverse dosing strategies and infusion modes.

Results

fAUC/MICCAZ/AVI was the most predictive index for avibactam against Enterobacteriaceae in both mice and humans, regardless of infusion mode. Against Pseudomonas aeruginosa, fT > CT predicted efficacy in mice, while fAUC/MICCAZ/AVI and fCmax/MICCAZ/AVI were more predictive in humans, particularly for continuous infusion regimens. Higher PTAs were achieved with increased avibactam doses relative to ceftazidime, particularly with 1:1 and 2:1 ceftazidime:avibactam ratios. Continuous infusion improved PTA against P. aeruginosa but had limited impact on Enterobacteriaceae.

Conclusion

The PK/PD indices predictive of avibactam efficacy varied by species (mice and humans), bacterial strains, and mode of infusion. Dosing simulations suggest that increasing avibactam relative to ceftazidime and using continuous infusion regimens may enhance bacterial killing. These findings highlight the importance of refining dosing strategies for both components of the combination therapy.

Place, publisher, year, edition, pages
Oxford University Press, 2025
National Category
Pharmaceutical Sciences
Identifiers
urn:nbn:se:uu:diva-551649 (URN)10.1093/jacamr/dlaf036 (DOI)001443985200001 ()40070893 (PubMedID)
Available from: 2025-02-27 Created: 2025-02-27 Last updated: 2025-03-26Bibliographically approved
Zhao, C., van den Berg, S., Wang, Z., Olsson, A., Aranzana-Climent, V., Malmberg, C., . . . Friberg, L. E. (2025). An integrative and translational PKPD modelling approach to explore the combined effect of polymyxin B and minocycline against Klebsiella pneumoniae. International Journal of Antimicrobial Agents, 65(3), Article ID 107443.
Open this publication in new window or tab >>An integrative and translational PKPD modelling approach to explore the combined effect of polymyxin B and minocycline against Klebsiella pneumoniae
Show others...
2025 (English)In: International Journal of Antimicrobial Agents, ISSN 0924-8579, E-ISSN 1872-7913, Vol. 65, no 3, article id 107443Article in journal (Refereed) Published
Abstract [en]

Objectives:

To expand a translational pharmacokinetic-pharmacodynamic (PKPD) modelling approach for assessing the combined effect of polymyxin B and minocycline against Klebsiella pneumoniae.

Methods:

A PKPD model developed based on in vitro static time-kill experiments of one strain (ARU613) was first translated to characterize that of a more susceptible strain (ARU705), and thereafter to dynamic time-kill experiments (both strains) and to a murine thigh infection model (ARU705 only). The PKPD model was updated stepwise using accumulated data. Predictions of bacterial killing in humans were performed.

Results:

The same model structure could be used in each translational step, with parameters being re- estimated. Dynamic data were well predicted by static-data-based models. The in vitro/in vivo differences were primarily quantified as a change in polymyxin B effect: a lower killing rate constant in vivo compared with in vitro (concentration of 3 mg/L corresponds to 0.05/h and 57/h, respectively), and a slower adaptive resistance rate (the constant in vivo was 2.5% of that in vitro ). There was no significant difference in polymyxin B-minocycline interaction functions. Predictions based on both in vitro and in vivo parameters indicated that the combination has a greater-than-monotherapy antibacterial effect in humans, forecasting a reduction of approximately 5 and 2 log10 colony-forming units/mL at 24 h, respectively, under combined therapy, while the maximum bacterial load was reached in monotherapy.

Conclusions:

This study demonstrated the utility of the PKPD modelling approach to understand translation of antibiotic effects across experimental systems, and showed a promising antibacterial effect of polymyxin B and minocycline in combination against K. pneumoniae.

Place, publisher, year, edition, pages
Elsevier, 2025
Keywords
Translational, pharmacokinetic-pharmacodynamic, modelling, Semi-mechanistic, pharmacokinetic-pharmacodynamic model, Antibiotic combination, Polymyxin B, Minocycline
National Category
Pharmaceutical Sciences Infectious Medicine
Identifiers
urn:nbn:se:uu:diva-551746 (URN)10.1016/j.ijantimicag.2025.107443 (DOI)001423872700001 ()39793934 (PubMedID)2-s2.0-85216961395 (Scopus ID)
Funder
Swedish Research Council, 2018-03296Swedish Research Council, 2019-05911Swedish Research Council, 2020-02320Vinnova, 2021-02699
Available from: 2025-03-26 Created: 2025-03-26 Last updated: 2025-03-26Bibliographically approved
O'Jeanson, A., Ioannidis, K., Nielsen, E. I., Galani, L., Ginosyan, A., Paskalis, H., . . . Karaiskos, I. (2025). Ceftazidime-avibactam (CAZ-AVI) pharmacokinetics in critically ill patients undergoing continuous venovenous hemodiafiltration (CVVHDF). International Journal of Antimicrobial Agents, 65(1), Article ID 107394.
Open this publication in new window or tab >>Ceftazidime-avibactam (CAZ-AVI) pharmacokinetics in critically ill patients undergoing continuous venovenous hemodiafiltration (CVVHDF)
Show others...
2025 (English)In: International Journal of Antimicrobial Agents, ISSN 0924-8579, E-ISSN 1872-7913, Vol. 65, no 1, article id 107394Article in journal (Refereed) Published
Abstract [en]

Purpose: To investigate the pharmacokinetics (PK) of ceftazidime-avibactam (CAZ-AVI) in critically ill patients undergoing continuous venovenous hemodiafiltration (CVVHDF), and compare with a general phase III trial population.

Methods: A prospective PK study was conducted in critically ill patients who received CVVHDF for acute kidney injury, treated with CAZ-AVI (1000/250 mg or 2000/500 mg q8h). Plasma and CVVHDF-circuit samples were collected to determine CAZ-AVI concentrations. Individual PK parameters at steady-state were estimated using non-compartmental analysis. For visual comparison, plasma concentrations from CVVHDF patients were overlaid with simulated data from patients not receiving CVVHDF based on previously developed population PK models.

Results: A total of 35 plasma samples and 16 CVVHDF-circuit samples were obtained from four patients, with two patients sampled on two separate occasions. Median total clearance and volume of distribution were 4.54 L/h and 73.2 L for CAZ and 10.5 L/h and 102 L for AVI, respectively. Median contribution of CVVHDF to total clearance was 19.8% for CAZ and 5.3% for AVI. Observed CAZ-AVI PK profiles were generally within the 90% confidence interval of model predictions, but the observed concentrations were notably lower early (0-2 h) and higher later (4-8 h) in the dosing interval, suggesting a higher volume of distribution.

Conclusions: These results suggest that the CAZ-AVI dose regimens used in this study can be applicable in critically ill patients undergoing CVVHDF, despite the different shape of the PK profiles observed in this population. Further research with a larger patient cohort is warranted to validate and refine these findings.

Place, publisher, year, edition, pages
Elsevier, 2025
Keywords
Ceftazidime-avibactam, CAZ-AVI, Pharmacokinetics, PK, Critically ill patients, Intensive care unit, ICU, Renal replacement therapy, RRT, Continuous venovenous, hemodiafiltration, CVVHDF
National Category
Pharmaceutical Sciences
Identifiers
urn:nbn:se:uu:diva-545152 (URN)10.1016/j.ijantimicag.2024.107394 (DOI)001386331100001 ()39581557 (PubMedID)2-s2.0-85211985518 (Scopus ID)
Funder
Swedish Research Council, 2022-00657EU, Horizon 2020, 861323
Note

De två sista författarna delar sistaförfattarskapet

Available from: 2024-12-12 Created: 2024-12-12 Last updated: 2025-03-03Bibliographically approved
Liu, H., Ibrahim, E. I. .., Centanni, M., Sarr, C., Venkatakrishnan, K. & Friberg, L. E. (2025). Integrated modeling of biomarkers, survival and safety in clinical oncology drug development. Advanced Drug Delivery Reviews, 216, Article ID 115476.
Open this publication in new window or tab >>Integrated modeling of biomarkers, survival and safety in clinical oncology drug development
Show others...
2025 (English)In: Advanced Drug Delivery Reviews, ISSN 0169-409X, E-ISSN 1872-8294, Vol. 216, article id 115476Article in journal (Refereed) Published
Abstract [en]

Model-based approaches, including population pharmacokinetic-pharmacodynamic modeling, have become an essential component in the clinical phases of oncology drug development. Over the past two decades, models have evolved to describe the temporal dynamics of biomarkers and tumor size, treatment-related adverse events, and their links to survival. Integrated models, defined here as models that incorporate at least two pharmacodynamic/ outcome variables, are applied to answer drug development questions through simulations, e.g., to support the exploration of alternative dosing strategies and study designs in subgroups of patients or other tumor indications. It is expected that these pharmacometric approaches will be expanded as regulatory authorities place further emphasis on early and individualized dosage optimization and inclusive patient-focused development strategies. This review provides an overview of integrated models in the literature, examples of the considerations that need to be made when applying these advanced pharmacometric approaches, and an outlook on the expected further expansion of model-informed drug development of anticancer drugs.Keywords: Anticancer drugs; Dose individualization; Dose optimization; Joint models; Model-informed drug development; Pharmacodynamic; Pharmacokinetic; Pharmacometrics; Tumor growth inhibition model.

Place, publisher, year, edition, pages
Elsevier, 2025
Keywords
Anticancer drugs, Pharmacokinetic, Pharmacodynamic, Pharmacometrics, Joint models, Model-informed drug developmen, t Dose optimization, Dose individualization, Tumor growth inhibition model
National Category
Pharmaceutical Sciences
Identifiers
urn:nbn:se:uu:diva-545150 (URN)10.1016/j.addr.2024.115476 (DOI)001371845700001 ()39577694 (PubMedID)2-s2.0-85210392122 (Scopus ID)
Funder
Swedish Cancer Society, 20 1226 PjFSwedish Cancer Society, 23 2921 Pj
Available from: 2024-12-12 Created: 2024-12-12 Last updated: 2025-01-09Bibliographically approved
Bahnasawy, S. M., Ahmed, H., Zeitlinger, M., Friberg, L. E. & Nielsen, E. I. (2025). Plasma effects on bacterial time-kill dynamics: Insights from a PK/PD modelling analysis. International Journal of Antimicrobial Agents, 65(2), Article ID 107441.
Open this publication in new window or tab >>Plasma effects on bacterial time-kill dynamics: Insights from a PK/PD modelling analysis
Show others...
2025 (English)In: International Journal of Antimicrobial Agents, ISSN 0924-8579, E-ISSN 1872-7913, Vol. 65, no 2, article id 107441Article in journal (Refereed) Published
Abstract [en]

In vitro time-kill curve (TKC) experiments are an important part of the pharmacokinetic- pharmacodynamic (PKPD) characterisation of antibiotics. Traditional TKCs use Mueller-Hinton broth (MHB), which lacks specific plasma components that could potentially influence the bacterial growth and killing dynamics, and affect translation to in vivo. This study aimed to evaluate the impact of plasma on the PKPD characterisation of two antibiotics; cefazolin and clindamycin. TKC experiments were conducted in pure MHB, and MHB spiked with 20% and 70% human plasma. Plasma protein binding (PPB) data were available, and a linear model described cefazolin's PPB, while clindamycin's PPB was best described by a second-order polynomial model. PKPD models were developed based on pure MHB and described drug effects using an Emax model, with consideration of adaptive resistance for cefazolin. The observed bacterial growth and killing in the plasma-spiked MHB TKC data was insufficiently described when applying the developed PPB and PKPD models. In plasma spiked MHB, a growth delay was observed, estimated to 0.25 h (20% plasma), or 2.90 h (70% plasma) for cefazolin, and 0.64 h (20% plasma), or 1.40 h (70% plasma) for clindamycin. Furthermore, the drug effect was higher than expected in plasma-spiked MHB, with bacterial stasis and/or killing at unbound concentrations below MIC, necessitating drug effect parameter scaling (C50 for cefazolin, Hill coefficient for clindamycin). The findings highlight significant differences in bacterial growth and killing dynamics between pure MHB and plasma-spiked MHB and exemplify how PKPD modelling may be used to improve the translation of in vitro results.

Place, publisher, year, edition, pages
Elsevier, 2025
Keywords
Time-kill curve (TKC), Pharmacokinetics-pharmacodynamics (PKPD), Plasma protein binding (PPB)
National Category
Pharmaceutical Sciences
Identifiers
urn:nbn:se:uu:diva-550400 (URN)10.1016/j.ijantimicag.2024.107441 (DOI)001409631200001 ()39778755 (PubMedID)2-s2.0-85215546161 (Scopus ID)
Funder
EU, Horizon 2020, 861323
Available from: 2025-02-19 Created: 2025-02-19 Last updated: 2025-03-18Bibliographically approved
Saporta, R., Madan, M. & Friberg, L. E. (2025). Simulation-based evaluation of the impact of dose fractionation study design on antibiotic PKPD analyses. JAC - Antimicrobial Resistance, 7(2), Article ID dlaf057.
Open this publication in new window or tab >>Simulation-based evaluation of the impact of dose fractionation study design on antibiotic PKPD analyses
2025 (English)In: JAC - Antimicrobial Resistance, E-ISSN 2632-1823, Vol. 7, no 2, article id dlaf057Article in journal (Refereed) Published
Abstract [en]

Objectives

To evaluate the impact of antibiotic dose fractionation study design on pharmacokinetic/pharmacodynamic (PK/PD) indices and PKPD model estimation.

Methods

PKPD models for meropenem and polymyxin B (PMB) were applied to (i) simulate various dose fractionation studies in mice to derive PK/PD indices and efficacy targets and (ii) perform stochastic simulations and estimations evaluating which efficacy assessment times, in addition to 24 h, would improve the estimation of drug effect parameters.

Results

The R2 values of PK/PD indices were primarily influenced by reductions of the dosing intervals for meropenem and by decreases of the lowest total daily dose for PMB. For certain study designs (e.g. frequent administration of higher meropenem doses), R2 values for fT > MIC and fAUC/MIC were similar. Efficacy target magnitudes were also sensitive to the selected doses. Additional efficacy assessment times improved parameter accuracy (e.g. 40% reduction in relative root mean squared error of PMB effect slope). The model parameter accuracy was more affected by the selection of time points for meropenem, which included resistance, than for PMB. Efficacy measurements in the first hours after treatment start (e.g. 2 and 6 h), in addition to 24 h, were essential for resistance characterization.

Conclusions

The choice of doses and fractionations impacted PK/PD index selection and efficacy target magnitude. Depending on the antibiotic, the dose or fractionation selection appeared to be the most critical. Early treatment efficacy measurements were beneficial to PKPD model-based analyses, particularly to describe resistance processes.

Place, publisher, year, edition, pages
Oxford University Press, 2025
National Category
Pharmaceutical Sciences
Identifiers
urn:nbn:se:uu:diva-549465 (URN)10.1093/jacamr/dlaf057 (DOI)001465438900001 ()40224358 (PubMedID)
Funder
National Academic Infrastructure for Supercomputing in Sweden (NAISS)Swedish Research Council, 2022-06725Swedish Research Council, 2022-00657EU, Horizon 2020, 861323
Note

De två första författarna delar förstaförfattarskapet

Available from: 2025-02-04 Created: 2025-02-04 Last updated: 2025-04-29Bibliographically approved
Ibrahim, E. I. K., Ellingsen, E. B., Mangsbo, S. M. & Friberg, L. (2024). Bridging responses to a human telomerase reverse transcriptase-based peptide cancer vaccine candidate in a mechanism-based model. International Immunopharmacology, 126, Article ID 111225.
Open this publication in new window or tab >>Bridging responses to a human telomerase reverse transcriptase-based peptide cancer vaccine candidate in a mechanism-based model
2024 (English)In: International Immunopharmacology, ISSN 1567-5769, E-ISSN 1878-1705, Vol. 126, article id 111225Article in journal (Refereed) Published
Abstract [en]

Therapeutic cancer vaccines are novel immuno-therapeutics, aiming to improve clinical outcomes with other immunotherapies. However, obstacles to their successful clinical development remain, which model-informed drug development approaches may address. UV1 is a telomerase based therapeutic cancer vaccine candidate being investigated in phase I clinical trials for multiple indications. We developed a mechanism-based model structure, using a nonlinear mixed‐effects modeling techniques, based on longitudinal tumor sizes (sum of the longest diameters, SLD), UV1-specific immunological assessment (stimulation index, SI) and overall survival (OS) data obtained from a UV1 phase I trial including non-small cell lung cancer (NSCLC) patients and a phase I/IIa trial including malignant melanoma (MM) patients. The final structure comprised a mechanistic tumor growth dynamics (TGD) model, a model describing the probability of observing a UV1-specific immune response (SI ≥ 3) and a time-to-event model for OS. The mechanistic TGD model accounted for the interplay between the vaccine peptides, immune system and tumor. The model-predicted UV1-specific effector CD4+ T cells induced tumor shrinkage with half-lives of 103 and 154 days in NSCLC and MM patients, respectively. The probability of observing a UV1-specific immune response was mainly driven by the model-predicted UV1-specific effector and memory CD4+ T cells. A high baseline SLD and a high relative increase from nadir were identified as main predictors for a reduced OS in NSCLC and MM patients, respectively. Our model predictions highlighted that additional maintenance doses, i.e. UV1 administration for longer periods, may result in more sustained tumor size shrinkage.

Place, publisher, year, edition, pages
Elsevier, 2024
Keywords
Therapeutic cancer vaccine, Immunotherapy, Model -informed drug development, Quantitative system pharmacology, Tumor growth dynamics model, Pharmacometric modelling framework
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:uu:diva-528375 (URN)10.1016/j.intimp.2023.111225 (DOI)001161913400001 ()37988911 (PubMedID)
Funder
Swedish Cancer Society, 20 1226 PjFSwedish Research Council, 2022-06725Swedish Research Council, 2018-05973National Academic Infrastructure for Supercomputing in Sweden (NAISS)Swedish National Infrastructure for Computing (SNIC)UPPMAX
Available from: 2024-05-21 Created: 2024-05-21 Last updated: 2024-05-21Bibliographically approved
Sanchez, J., Claus, C., McIntyre, C., Tanos, T., Boehnke, A., Friberg, L. E., . . . Frances, N. (2024). Combining mathematical modeling, in vitro data and clinical target expression to support bispecific antibody binding affinity selection: a case example with FAP-4-1BBL. Frontiers in Pharmacology, 15, Article ID 1472662.
Open this publication in new window or tab >>Combining mathematical modeling, in vitro data and clinical target expression to support bispecific antibody binding affinity selection: a case example with FAP-4-1BBL
Show others...
2024 (English)In: Frontiers in Pharmacology, E-ISSN 1663-9812, Vol. 15, article id 1472662Article in journal (Refereed) Published
Abstract [en]

The majority of bispecific costimulatory antibodies in cancer immunotherapy are capable of exerting tumor-specific T-cell activation by simultaneously engaging both tumor-associated targets and costimulatory receptors expressed by T cells. The amount of trimeric complex formed when the bispecific antibody is bound simultaneously to the T cell receptor and the tumor-associated target follows a bell-shaped curve with increasing bispecific antibody exposure/dose. The shape of the curve is determined by the binding affinities of the bispecific antibody to its two targets and target expression. Here, using the case example of FAP-4-1BBL, a fibroblast activation protein alpha (FAP)-directed 4-1BB (CD137) costimulator, the impact of FAP-binding affinity on trimeric complex formation and pharmacology was explored using mathematical modeling and simulation. We quantified (1) the minimum number of target receptors per cell required to achieve pharmacological effect, (2) the expected coverage of the patient population for 19 different solid tumor indications, and (3) the range of pharmacologically active exposures as a function of FAP-binding affinity. A 10-fold increase in FAP-binding affinity (from a dissociation constant [KD] of 0.7 nM–0.07 nM) was predicted to reduce the number of FAP receptors needed to achieve 90% of the maximum pharmacological effect from 13,400 to 4,000. Also, the number of patients with colon cancer that would achieve 90% of the maximum effect would increase from 6% to 39%. In this work, a workflow to select binding affinities for bispecific antibodies that integrates preclinical in vitro data, mathematical modeling and simulation, and knowledge on target expression in the patient population, is provided. The early implementation of this approach can increase the probability of success with cancer immunotherapy in clinical development.

Place, publisher, year, edition, pages
Frontiers Media S.A., 2024
Keywords
immunotherapy, bispecific antibody, modeling, simulation, binding affinity, oncology, pharmacodynamics, FAP-4-1BBL
National Category
Cancer and Oncology
Identifiers
urn:nbn:se:uu:diva-542797 (URN)10.3389/fphar.2024.1472662 (DOI)001340381000001 ()39444607 (PubMedID)
Available from: 2024-11-15 Created: 2024-11-15 Last updated: 2024-12-10Bibliographically approved
Centanni, M., Nijhuis, J., Karlsson, M. O. & Friberg, L. E. (2024). Comparative Analysis of Traditional and Pharmacometric-Based Pharmacoeconomic Modeling in the Cost-Utility Evaluation of Sunitinib Therapy. PharmacoEconomics (Auckland)
Open this publication in new window or tab >>Comparative Analysis of Traditional and Pharmacometric-Based Pharmacoeconomic Modeling in the Cost-Utility Evaluation of Sunitinib Therapy
2024 (English)In: PharmacoEconomics (Auckland), ISSN 1170-7690, E-ISSN 1179-2027Article in journal (Refereed) Epub ahead of print
Abstract [en]

Background: Cost-utility analyses (CUAs) increasingly use models to predict long-term outcomes and translate trial data to real-world settings. Model structure uncertainty affects these predictions. This study compares pharmacometric against traditional pharmacoeconomic model evaluations for CUAs of sunitinib in gastrointestinal stromal tumors (GIST).

Methods: A two-arm trial comparing sunitinib 37.5 mg daily with no treatment was simulated using a pharmacometric-based pharmacoeconomic model framework. Overall, four existing models [time-to-event (TTE) and Markov models] were re-estimated to the survival data and linked to logistic regression models describing the toxicity data [neutropenia, thrombocytopenia, hypertension, fatigue, and hand-foot syndrome (HFS)] to create traditional pharmacoeconomic model frameworks. All five frameworks were used to simulate clinical outcomes and sunitinib treatment costs, including a therapeutic drug monitoring (TDM) scenario.

Results: The pharmacometric model framework predicted that sunitinib treatment costs an additional 142,756 euros per quality adjusted life year (QALY) compared with no treatment, with deviations - 21.2% (discrete Markov), - 15.1% (continuous Markov), + 7.2% (TTE Weibull), and + 39.6% (TTE exponential) from the traditional model frameworks. The pharmacometric framework captured the change in toxicity over treatment cycles (e.g., increased HFS incidence until cycle 4 with a decrease thereafter), a pattern not observed in the pharmacoeconomic frameworks (e.g., stable HFS incidence over all treatment cycles). Furthermore, the pharmacoeconomic frameworks excessively forecasted the percentage of patients encountering subtherapeutic concentrations of sunitinib over the course of time (pharmacoeconomic: 24.6% at cycle 2 to 98.7% at cycle 16, versus pharmacometric: 13.7% at cycle 2 to 34.1% at cycle 16).

Conclusions: Model structure significantly influences CUA predictions. The pharmacometric-based model framework more closely represented real-world toxicity trends and drug exposure changes. The relevance of these findings depends on the specific question a CUA seeks to address.

Place, publisher, year, edition, pages
Springer, 2024
National Category
Pharmacology and Toxicology
Identifiers
urn:nbn:se:uu:diva-545158 (URN)10.1007/s40273-024-01438-z (DOI)001321553500001 ()2-s2.0-85205035560 (Scopus ID)
Funder
Swedish Cancer Society, CAN 23 2921 PjSwedish Cancer Society, CAN 20 1226 PjFUppsala University
Available from: 2024-12-12 Created: 2024-12-12 Last updated: 2024-12-17
Minichmayr, I. & Friberg, L. (2024). Impact of continuous-infusion meropenem degradation and infusion bag changes on bacterial killing of Pseudomonas aeruginosa based on model-informed translation. International Journal of Antimicrobial Agents, 64(2), Article ID 107236.
Open this publication in new window or tab >>Impact of continuous-infusion meropenem degradation and infusion bag changes on bacterial killing of Pseudomonas aeruginosa based on model-informed translation
2024 (English)In: International Journal of Antimicrobial Agents, ISSN 0924-8579, E-ISSN 1872-7913, Vol. 64, no 2, article id 107236Article in journal (Refereed) Published
Abstract [en]

Background

Continuous infusion of meropenem has been proposed to increase target attainment in critically ill patients, although stability might limit its practical use. This study investigated the impact of meropenem degradation and infusion bag changes on the concentration-time profiles and bacterial growth and killing of P. aeruginosa given different continuous-infusion solutions.

Methods

A semi-mechanistic pharmacokinetic-pharmacodynamic (PK-PD) model quantifying meropenem concentrations (CMEM) and bacterial counts of a resistant P. aeruginosa strain (ARU552, MIC = 16 mg/L) over 24 h was used to translate in vitro antibiotic effects to patients with severe infections. Concentration-dependent drug degradation of saline infusion solutions was considered using an additional compartment in the population PK model. CMEM, fT>MIC (time that concentrations exceed the MIC) and total bacterial load (BTOT) after 24 h were simulated for different scenarios (n = 144), considering low- and high-dose regimens (3000/6000 mg/day±loading dose), clinically relevant infusion solutions (20/40/50 mg/mL), different intervals of infusion bag changes (every 8/24 h, q8/24 h), and varied renal function (creatinine clearance 40/80/120 mL/min) and MIC values (8/16 mg/L).

Results

Highest deviations between changing infusion bags q8h and q24h were observed for 50 mg/mL solutions and scenarios with CMEM_24h close to the MIC, with differences (Δ) in CMEM_24h up to 4.9 mg/L, ΔfT>MIC≤65.7%, and ΔBTOT_24h≤1.1 log10 CFU/mL, thus affecting conclusions on whether bacteriostasis was reached.

Conclusions

In summary, this study indicated that for continuous infusion of meropenem, eight-hourly infusion bag changes improved PK/PD target attainment and might be beneficial particularly for high meropenem concentrations of saline infusion solutions and for plasma concentrations in close proximity to the MIC.

Place, publisher, year, edition, pages
Elsevier, 2024
Keywords
Meropenem, Continuous infusion, Stability, PKPD, Translation
National Category
Infectious Medicine Pharmaceutical Sciences
Identifiers
urn:nbn:se:uu:diva-535405 (URN)10.1016/j.ijantimicag.2024.107236 (DOI)001266928700001 ()38851463 (PubMedID)
Funder
Swedish Research Council, 2018-03296
Available from: 2024-07-30 Created: 2024-07-30 Last updated: 2024-07-30Bibliographically approved
Projects
Developing combinations of CO-ACTIVE antimicrobials and non-antimicrobials [2015-06826_VR]; Uppsala UniversityA pharmacometric framework for model-based translation of antibiotic effects in vitro to outcomes in patients infected by resistant bacteria [2018-03296_VR]; Uppsala University; Publications
Zhao, C., Kristoffersson, A. N., Khan, D. D., Lagerbäck, P., Lustig, U., Cao, S., . . . Friberg, L. E. (2024). Quantifying combined effects of colistin and ciprofloxacin against Escherichia coli in an in silico pharmacokinetic-pharmacodynamic model. Scientific Reports, 14(1), Article ID 11706. Thorsted, A., Pham, A. D., Friberg, L. E. & Nielsen, E. I. (2023). Model‐based assessment of neutrophil‐mediated phagocytosis and digestion of bacteria across in vitro and in vivo studies. CPT: Pharmacometrics and Systems Pharmacology (PSP), 12(12), 1972-1987
Enabling efficient drug development of antibiotic therapy against multi-resistant pathogens: Establishment of a modelling framework for translating response from rabbits to patients [2022-00657_VR]; Uppsala University; Publications
Saporta, R., Madan, M. & Friberg, L. E. (2025). Simulation-based evaluation of the impact of dose fractionation study design on antibiotic PKPD analyses. JAC - Antimicrobial Resistance, 7(2), Article ID dlaf057.
Organisations
Identifiers
ORCID iD: ORCID iD iconorcid.org/0000-0002-2979-679X

Search in DiVA

Show all publications